1
|
Malešević A, Tucović D, Kulaš J, Mirkov I, Popović D, Čakić Milošević M, Popov Aleksandrov A. Impact of Skin Exposure to Benzo[a]pyrene in Rat Model: Insights into Epidermal Cell Function and Draining Lymph Node Cell Response. Int J Mol Sci 2024; 25:8631. [PMID: 39201318 PMCID: PMC11354278 DOI: 10.3390/ijms25168631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 07/29/2024] [Accepted: 08/02/2024] [Indexed: 09/02/2024] Open
Abstract
The skin is a direct target of the air pollutant benzo[a]pyrene (BaP). While its carcinogenic qualities are well-studied, the immunotoxicity of BaP after dermal exposure is less understood. This study examines the immunomodulatory effects of a 10-day epicutaneous BaP application, in environmentally/occupationally relevant doses, by analyzing ex vivo skin immune response (skin explant, epidermal cells and draining lymph node/DLN cell activity), alongside the skin's reaction to sensitization with experimental hapten dinitrochlorobenzene (DNCB). The results show that BaP application disrupts the structure of the epidermal layer and promotes immune cell infiltration in the dermis. BaP exposure led to oxidative stress in epidermal cells, characterized by decreased reduced glutathione and increased AHR and Cyp1A1 expression. Production and gene expression of proinflammatory cytokines (TNF, IL-1β) by epidermal cells decreased, while IL-10 response increased. Decreased spontaneous production of IFN-γ and IL-17, along with unchanged IL-10, was observed in DLC cells, whereas ConA-stimulated production of these cytokines was elevated. Local immunosuppression caused by BaP application seems to reduce the skin's response to an additional stimulus, evidenced by decreased effector activity of DLN cells three days after sensitization with DNCB. These findings provide new insight into the immunomodulatory effects and health risks associated with skin exposure to BaP.
Collapse
Affiliation(s)
- Anastasija Malešević
- Immunotoxicology Group, Department of Ecology, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, 142 Bulevar Despota Stefana, 11000 Belgrade, Serbia; (A.M.); (D.T.); (J.K.); (I.M.); (D.P.)
| | - Dina Tucović
- Immunotoxicology Group, Department of Ecology, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, 142 Bulevar Despota Stefana, 11000 Belgrade, Serbia; (A.M.); (D.T.); (J.K.); (I.M.); (D.P.)
| | - Jelena Kulaš
- Immunotoxicology Group, Department of Ecology, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, 142 Bulevar Despota Stefana, 11000 Belgrade, Serbia; (A.M.); (D.T.); (J.K.); (I.M.); (D.P.)
| | - Ivana Mirkov
- Immunotoxicology Group, Department of Ecology, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, 142 Bulevar Despota Stefana, 11000 Belgrade, Serbia; (A.M.); (D.T.); (J.K.); (I.M.); (D.P.)
| | - Dušanka Popović
- Immunotoxicology Group, Department of Ecology, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, 142 Bulevar Despota Stefana, 11000 Belgrade, Serbia; (A.M.); (D.T.); (J.K.); (I.M.); (D.P.)
| | - Maja Čakić Milošević
- Institute of Zoology, Faculty of Biology, University of Belgrade, 16 Studentski trg, 11000 Belgrade, Serbia;
| | - Aleksandra Popov Aleksandrov
- Immunotoxicology Group, Department of Ecology, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, 142 Bulevar Despota Stefana, 11000 Belgrade, Serbia; (A.M.); (D.T.); (J.K.); (I.M.); (D.P.)
| |
Collapse
|
2
|
Ando M, Yamaguchi H, Morimoto A, Iwashita N, Takagi Y, Nagane M, Yoshinari T, Fukuyama T. Chronic oral exposure to low-concentration fumonisin B2 significantly exacerbates the inflammatory responses of allergies in mice via inhibition of IL-10 release by regulatory T cells in gut-associated lymphoid tissue. Arch Toxicol 2023; 97:2707-2719. [PMID: 37589943 DOI: 10.1007/s00204-023-03579-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 08/03/2023] [Indexed: 08/18/2023]
Abstract
Contamination with fumonisins produced by Fusarium spp. is rapidly growing in both developing and developed countries. The purpose of this study was to determine whether oral exposure to fumonisin contributed to the development of allergic diseases. We initially examined the immunotoxic potential of short-term, oral administration of fumonisin B1 (FB1, 1 mg/kg) and fumonisin B2 (FB2, 1 mg/kg), both naturally occurring fumonisins, using a BALB/c mouse model of allergic contact dermatitis and Dermatophagoides farina-induced asthma. Using an NC/nga mouse model of atopic dermatitis (AD), we evaluated the adverse effects of subchronic oral exposure to low concentrations of FB2 (2 or 200 μg/kg). Finally, we explored the influence of FB2 on regulatory T cell proliferation and function in mesenteric lymph nodes after 1-week oral exposure to FB2 in BALB/c mice. Oral exposure to FB2 markedly exacerbated the symptoms of allergy, including skin thickness, histological evaluation, immunocyte proliferation, and proinflammatory cytokine production, although no change was observed following exposure to FB1. Furthermore, oral exposure to low concentrations of FB2 considerably exacerbated the AD scores, skin thickness, transepidermal water loss, histological features, and proinflammatory cytokine production. The aggravated allergic symptoms induced by oral exposure to FB2 could be attributed to the direct inhibition of IL-10 production by regulatory T cells in mesenteric lymph nodes. Our findings indicate that the recommended maximum fumonisin level should be reconsidered based on the potential for allergy development.
Collapse
Affiliation(s)
- Mana Ando
- School of Veterinary Medicine, Laboratory of Veterinary Pharmacology, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara, Kanagawa, 252-5201, Japan
| | - Hiroki Yamaguchi
- School of Veterinary Medicine, Laboratory of Veterinary Pharmacology, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara, Kanagawa, 252-5201, Japan
| | - Ai Morimoto
- School of Veterinary Medicine, Laboratory of Veterinary Pharmacology, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara, Kanagawa, 252-5201, Japan
| | - Naoki Iwashita
- School of Veterinary Medicine, Laboratory of Veterinary Pharmacology, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara, Kanagawa, 252-5201, Japan
- Bioalch Co., Ltd., 3-28 Honshuku-cho, Fuchu, Tokyo, Japan
| | - Yoshiichi Takagi
- School of Veterinary Medicine, Laboratory of Veterinary Pharmacology, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara, Kanagawa, 252-5201, Japan
- Japan SLC, Inc, 85 Ohara-cho, Kita-ku, Hamamatsu, Shizuoka, Japan
| | - Masaki Nagane
- School of Veterinary Medicine, Laboratory of Veterinary Pharmacology, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara, Kanagawa, 252-5201, Japan
- Center for Human and Animal Symbiosis Science, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara, Kanagawa, Japan
| | - Tomoya Yoshinari
- Division of Microbiology, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, Japan
| | - Tomoki Fukuyama
- School of Veterinary Medicine, Laboratory of Veterinary Pharmacology, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara, Kanagawa, 252-5201, Japan.
- Center for Human and Animal Symbiosis Science, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara, Kanagawa, Japan.
| |
Collapse
|
3
|
Du B, Xiao X, Wang H, Li W, Xia Z, Yang P, Huang SK, Yuan R, Liu J, Han M, Zou Y, Zhu J, He D, Lyu J, Jin X, Xu X, Wang J, Yang H, Xiao L, Liu X, Kristiansen K. Evaluation of the Impact of BaP Exposure on the Gut Microbiota and Allergic Responses in an OVA-Sensitized Mouse Model. ENVIRONMENTAL HEALTH PERSPECTIVES 2023; 131:67004. [PMID: 37267060 DOI: 10.1289/ehp11874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
BACKGROUND Exposure to environmental pollutants, including benzo[a]pyrene (BaP), has been implicated in allergic diseases and intestinal microbiota homeostasis, but the environment-microbiota-immunity triangular relationship and to what extent BaP-induced remodeling of the gut microbiota contributes to intestinal allergic inflammation remain to be established. OBJECTIVES We investigated the impact of BaP on intestinal allergic inflammation and examined the relationship between this effect and gut microbiota dysbiosis. We explored the potential ability of intestinal bacteria to degrade BaP and alleviate cytotoxicity as a detoxification strategy to counteract the effects of BaP exposure. METHODS We combined microbiome shotgun metagenomics with animal histological and intestinal allergic inflammatory responses to assess the effects of BaP (50μg/mouse per day) in a 23-d toxicity test in antigen-induced allergic female mice. In addition, genome annotation, quantitative analysis of BaP, and in vitro cytotoxicity-tests using CaCo-2 cells were conducted to infer the role of intestinal bacteria in BaP detoxification. RESULTS BaP exposure impacted the taxonomic composition and the functional potential of the gut microbiota and aggravated antigen-induced intestinal allergic inflammatory responses. The level of inflammatory cytokines correlated with the abundance of specific bacterial taxa, including Lachnospiraceae bacterium 28-4 and Alistipes inops. We identified 614 bacteria harboring genes implicated in the degradation of BaP, and 4 of these bacterial strains were shown to significantly reduce the cytotoxicity of BaP to CaCo-2 cells in vitro. DISCUSSION Using allergic female mice as a model, we investigated the relationship between BaP, microbiota, and host immune reactions, highlighting the role of gut bacteria in BaP-aggravated allergic reactions. Our findings offer novel insight toward establishing the causal relationship between BaP exposure and the occurrence of allergic disorders. Identifying gut bacteria that degrade BaP may provide new strategies for ameliorating BaP cytotoxicity. https://doi.org/10.1289/EHP11874.
Collapse
Affiliation(s)
- Beibei Du
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI-Shenzhen, Shenzhen, China
| | - Xiaojun Xiao
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen Key Laboratory of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Huailing Wang
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen Key Laboratory of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Wenxi Li
- BGI-Shenzhen, Shenzhen, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | | | - Pingchang Yang
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen Key Laboratory of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
- National Institute of Environmental Health Sciences, National Health Research Institutes, Taiwan, China
| | - Shau-Ku Huang
- National Institute of Environmental Health Sciences, National Health Research Institutes, Taiwan, China
- Department of Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
- Department of Medicine, Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ruyi Yuan
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen Key Laboratory of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Jie Liu
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen Key Laboratory of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Mo Han
- BGI-Shenzhen, Shenzhen, China
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
- Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, Shenzhen, China
| | - Yuanqiang Zou
- BGI-Shenzhen, Shenzhen, China
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
- Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, Shenzhen, China
- Qingdao-Europe Advanced Institute for Life Sciences, Qingdao, China
| | | | | | | | - Xin Jin
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI-Shenzhen, Shenzhen, China
| | - Xun Xu
- BGI-Shenzhen, Shenzhen, China
| | | | - Huanming Yang
- BGI-Shenzhen, Shenzhen, China
- Qingdao-Europe Advanced Institute for Life Sciences, Qingdao, China
| | - Liang Xiao
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI-Shenzhen, Shenzhen, China
- Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, Shenzhen, China
- Qingdao-Europe Advanced Institute for Life Sciences, Qingdao, China
| | - Xiaoyu Liu
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen Key Laboratory of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Karsten Kristiansen
- BGI-Shenzhen, Shenzhen, China
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
- Qingdao-Europe Advanced Institute for Life Sciences, Qingdao, China
| |
Collapse
|
4
|
Schmidt JR, Haupt J, Riemschneider S, Kämpf C, Löffler D, Blumert C, Reiche K, Koehl U, Kalkhof S, Lehmann J. Transcriptomic signatures reveal a shift towards an anti-inflammatory gene expression profile but also the induction of type I and type II interferon signaling networks through aryl hydrocarbon receptor activation in murine macrophages. Front Immunol 2023; 14:1156493. [PMID: 37287978 PMCID: PMC10242070 DOI: 10.3389/fimmu.2023.1156493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 05/09/2023] [Indexed: 06/09/2023] Open
Abstract
Introduction The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that regulates a broad range of target genes involved in the xenobiotic response, cell cycle control and circadian rhythm. AhR is constitutively expressed in macrophages (Mϕ), acting as key regulator of cytokine production. While proinflammatory cytokines, i.e., IL-1β, IL-6, IL-12, are suppressed through AhR activation, anti-inflammatory IL-10 is induced. However, the underlying mechanisms of those effects and the importance of the specific ligand structure are not yet completely understood. Methods Therefore, we have compared the global gene expression pattern in activated murine bone marrow-derived macrophages (BMMs) subsequently to exposure with either benzo[a]pyrene (BaP) or indole-3-carbinol (I3C), representing high-affinity vs. low-affinity AhR ligands, respectively, by means of mRNA sequencing. AhR dependency of observed effects was proved using BMMs from AhR-knockout (Ahr-/-) mice. Results and discussion In total, more than 1,000 differentially expressed genes (DEGs) could be mapped, covering a plethora of AhR-modulated effects on basal cellular processes, i.e., transcription and translation, but also immune functions, i.e., antigen presentation, cytokine production, and phagocytosis. Among DEGs were genes that are already known to be regulated by AhR, i.e., Irf1, Ido2, and Cd84. However, we identified DEGs not yet described to be AhR-regulated in Mϕ so far, i.e., Slpi, Il12rb1, and Il21r. All six genes likely contribute to shifting the Mϕ phenotype from proinflammatory to anti-inflammatory. The majority of DEGs induced through BaP were not affected through I3C exposure, probably due to higher AhR affinity of BaP in comparison to I3C. Mapping of known aryl hydrocarbon response element (AHRE) sequence motifs in identified DEGs revealed more than 200 genes not possessing any AHRE, and therefore being not eligible for canonical regulation. Bioinformatic approaches modeled a central role of type I and type II interferons in the regulation of those genes. Additionally, RT-qPCR and ELISA confirmed a AhR-dependent expressional induction and AhR-dependent secretion of IFN-γ in response to BaP exposure, suggesting an auto- or paracrine activation pathway of Mϕ.
Collapse
Affiliation(s)
- Johannes R. Schmidt
- Department of Preclinical Development and Validation, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Leipzig, Germany
| | - Janine Haupt
- Department of Preclinical Development and Validation, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Leipzig, Germany
| | - Sina Riemschneider
- Department of Preclinical Development and Validation, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Christoph Kämpf
- Department of Diagnostics, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Dennis Löffler
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Leipzig, Germany
- Department of Diagnostics, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Conny Blumert
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Leipzig, Germany
- Department of Diagnostics, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Kristin Reiche
- Department of Diagnostics, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
- Institute for Clinical Immunology, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Ulrike Koehl
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Leipzig, Germany
- Institute for Clinical Immunology, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Stefan Kalkhof
- Department of Preclinical Development and Validation, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Leipzig, Germany
- Department of Applied Sciences, Institute for Bioanalysis, Coburg University of Applied Sciences and Arts, Coburg, Germany
| | - Jörg Lehmann
- Department of Preclinical Development and Validation, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Leipzig, Germany
| |
Collapse
|
5
|
Perdew GH, Esser C, Snyder M, Sherr DH, van den Bogaard EH, McGovern K, Fernández-Salguero PM, Coumoul X, Patterson AD. The Ah Receptor from Toxicity to Therapeutics: Report from the 5th AHR Meeting at Penn State University, USA, June 2022. Int J Mol Sci 2023; 24:5550. [PMID: 36982624 PMCID: PMC10058801 DOI: 10.3390/ijms24065550] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/10/2023] [Indexed: 03/16/2023] Open
Abstract
The aryl hydrocarbon receptor (AHR) is a sensor of low-molecular-weight molecule signals that originate from environmental exposures, the microbiome, and host metabolism. Building upon initial studies examining anthropogenic chemical exposures, the list of AHR ligands of microbial, diet, and host metabolism origin continues to grow and has provided important clues as to the function of this enigmatic receptor. The AHR has now been shown to be directly involved in numerous biochemical pathways that influence host homeostasis, chronic disease development, and responses to toxic insults. As this field of study has continued to grow, it has become apparent that the AHR is an important novel target for cancer, metabolic diseases, skin conditions, and autoimmune disease. This meeting attempted to cover the scope of basic and applied research being performed to address possible applications of our basic knowledge of this receptor on therapeutic outcomes.
Collapse
Affiliation(s)
- Gary H. Perdew
- Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, Penn State University, University Park, PA 16802, USA
| | - Charlotte Esser
- IUF-Leibniz Research Institute for Environmental Medicine, Auf’m Hennekamp 50, 40225 Düsseldorf, Germany
| | - Megan Snyder
- Department of Environmental Health, Boston University School of Public Health, 72 East Concord Street, Boston, MA 02118, USA
| | - David H. Sherr
- Department of Environmental Health, Boston University School of Public Health, 72 East Concord Street, Boston, MA 02118, USA
| | - Ellen H. van den Bogaard
- Department of Dermatology, Radboud University Medical Center, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Karen McGovern
- Ikena Oncology, Inc., 645 Summer Street Suite 101, Boston, MA 02210, USA
| | - Pedro M. Fernández-Salguero
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, Universidad de Extremadura, Avenida de Elvas s/n, 06071 Badajoz, Spain
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), Avenida de la Investigación s/n, 06071 Badajoz, Spain
| | - Xavier Coumoul
- INSERM UMR-S1124, 45 rue des Saints-Peères, 75006 Paris, France
| | - Andrew D. Patterson
- Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, Penn State University, University Park, PA 16802, USA
| |
Collapse
|
6
|
Liu KY, Gao Y, Xiao W, Fu J, Huang S, Han X, Hsu SH, Xiao X, Huang SK, Zhou Y. Multidimensional Analysis of Lung Lymph Nodes in a Mouse Model of Allergic Lung Inflammation following PM2.5 and Indeno[1,2,3- cd]pyrene Exposure. ENVIRONMENTAL HEALTH PERSPECTIVES 2023; 131:37014. [PMID: 36975775 PMCID: PMC10044348 DOI: 10.1289/ehp11580] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 01/23/2023] [Accepted: 02/09/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Ambient particulate matter with an aerodynamic diameter of ≤2.5 μm (PM2.5) is suggested to act as an adjuvant for allergen-mediated sensitization and recent evidence suggests the importance of T follicular helper (Tfh) cells in allergic diseases. However, the impact of PM2.5 exposure and its absorbed polycyclic aromatic hydrocarbon (PAHs) on Tfh cells and humoral immunity remains unknown. OBJECTIVES We aimed to explore the impact of environmental PM2.5 and indeno[1,2,3-cd]pyrene (IP), a prominent PAH, as a model, on Tfh cells and the subsequent pulmonary allergic responses. METHODS PM2.5- or IP-mediated remodeling of cellular composition in lung lymph nodes (LNs) was determined by mass cytometry in a house dust mite (HDM)-induced mouse allergic lung inflammation model. The differentiation and function of Tfh cells in vitro were analyzed by flow cytometry, quantitative reverse transcription polymerase chain reaction, enzyme-linked immunosorbent assay, chromatin immunoprecipitation, immunoprecipitation, and western blot analyses. RESULTS Mice exposed to PM2.5 during the HDM sensitization period demonstrated immune cell population shifts in lung LNs as compared with those sensitized with HDM alone, with a greater number of differentiated Tfh2 cells, enhanced allergen-induced immunoglobulin E (IgE) response and pulmonary inflammation. Similarly enhanced phenotypes were also found in mice exposed to IP and sensitized with HDM. Further, IP administration was found to induce interleukin-21 (Il21) and Il4 expression and enhance Tfh2 cell differentiation in vitro, a finding which was abrogated in aryl hydrocarbon receptor (AhR)-deficient CD4+ T cells. Moreover, we showed that IP exposure increased the interaction of AhR and cellular musculoaponeurotic fibrosarcoma (c-Maf) and its occupancy on the Il21 and Il4 promoters in differentiated Tfh2 cells. DISCUSSION These findings suggest that the PM2.5 (IP)-AhR-c-Maf axis in Tfh2 cells was important in allergen sensitization and lung inflammation, thus adding a new dimension in the understanding of Tfh2 cell differentiation and function and providing a basis for establishing the environment-disease causal relationship. https://doi.org/10.1289/EHP11580.
Collapse
Affiliation(s)
- Kwei-Yan Liu
- Department of Respirology & Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
- Institute of Pediatrics, Children’s Hospital of Fudan University, National Children’s Medical Center, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai, China
| | - Yajing Gao
- Institute of Pediatrics, Children’s Hospital of Fudan University, National Children’s Medical Center, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai, China
| | - Wenfeng Xiao
- Institute of Pediatrics, Children’s Hospital of Fudan University, National Children’s Medical Center, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai, China
| | - Jinrong Fu
- Institute of Pediatrics, Children’s Hospital of Fudan University, National Children’s Medical Center, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Department of General Medicine, Children’s Hospital of Fudan University, Shanghai, China
| | - Saihua Huang
- Institute of Pediatrics, Children’s Hospital of Fudan University, National Children’s Medical Center, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai, China
| | - Xiao Han
- Institute of Pediatrics, Children’s Hospital of Fudan University, National Children’s Medical Center, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai, China
| | - Shih-Hsien Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Xiaojun Xiao
- Institute of Allergy and Immunology, School of Medicine, Shenzhen University, Shenzhen, China
| | - Shau-Ku Huang
- Department of Respirology & Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
- Institute of Allergy and Immunology, School of Medicine, Shenzhen University, Shenzhen, China
- National Institute of Environmental Health Sciences, National Health Research Institutes, Taiwan
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yufeng Zhou
- Institute of Pediatrics, Children’s Hospital of Fudan University, National Children’s Medical Center, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Xu S, Zhou Y, Yu L, Huang X, Huang J, Wang K, Liu Z. Protective Effect of Eurotium cristatum Fermented Loose Dark Tea and Eurotium cristatum Particle on MAPK and PXR/AhR Signaling Pathways Induced by Electronic Cigarette Exposure in Mice. Nutrients 2022; 14:nu14142843. [PMID: 35889800 PMCID: PMC9318283 DOI: 10.3390/nu14142843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/29/2022] [Accepted: 07/02/2022] [Indexed: 02/01/2023] Open
Abstract
Electronic-cigarette smoke (eCS) has been shown to cause a degree of oxidative stress and inflammatory damage in lung tissue. The aim of this study was to evaluate the repair mechanism of Eurotium cristatum fermented loose dark tea (ECT) and Eurotium cristatum particle metabolites (ECP) sifted from ECT after eCS-induced injury in mice. Sixty C57BL/6 mice were randomly divided into a blank control group, an eCS model group, an eCS + 600 mg/kg ECP treatment group, an eCS + 600 mg/kg ECT treatment group, an eCS + 600 mg/kg ECP prevention group, and an eCS + 600 mg/kg ECT prevention group. The results show that ECP and ECT significantly reduced the eCS-induced oxidative stress and inflammation and improved histopathological changes in the lungs in mice with eCS-induced liver injury. Western blot analysis further revealed that ECP and ECT significantly inhibited the eCS-induced upregulation of the phosphorylation levels of the extracellular Regulated protein Kinases (ERK), c-Jun N-terminal kinase (JNK) and p38mitogen-activated protein kinases (p38MAPK) proteins, and significantly increased the eCS-induced downregulation of the expression levels of the pregnane X receptor (PXR) and aryl hydrocarbon receptor (AhR) proteins. Conclusively, these findings show that ECP and ECT have a significant repairing effect on the damage caused by eCS exposure through the MAPK and PXR/AhR signaling pathways; ECT has a better effect on preventing eCS-induced injury and is suitable as a daily healthcare drink; ECP has a better therapeutic effect after eCS-induced injury, and might be a potential therapeutic candidate for the treatment of eCS-induced injury.
Collapse
Affiliation(s)
- Shuai Xu
- Key Laboratory of Tea Science of Ministry of Education, National Research Center of Engineering Technology for Utilization of Functional Ingredients from Botanicals, College of Horticulture, Hunan Agricultural University, Changsha 410128, China; (S.X.); (Y.Z.); (X.H.); (J.H.); (K.W.)
| | - Yufei Zhou
- Key Laboratory of Tea Science of Ministry of Education, National Research Center of Engineering Technology for Utilization of Functional Ingredients from Botanicals, College of Horticulture, Hunan Agricultural University, Changsha 410128, China; (S.X.); (Y.Z.); (X.H.); (J.H.); (K.W.)
| | - Lijun Yu
- Key Laboratory of Tea Science of Ministry of Education, National Research Center of Engineering Technology for Utilization of Functional Ingredients from Botanicals, College of Horticulture, Hunan Agricultural University, Changsha 410128, China; (S.X.); (Y.Z.); (X.H.); (J.H.); (K.W.)
- Hunan Provincial Key Laboratory for Germplasm Innovation and Utilization of Crop, Hunan Agricultural University, Changsha 410128, China
- Correspondence: (L.Y.); (Z.L.)
| | - Xiangxiang Huang
- Key Laboratory of Tea Science of Ministry of Education, National Research Center of Engineering Technology for Utilization of Functional Ingredients from Botanicals, College of Horticulture, Hunan Agricultural University, Changsha 410128, China; (S.X.); (Y.Z.); (X.H.); (J.H.); (K.W.)
| | - Jianan Huang
- Key Laboratory of Tea Science of Ministry of Education, National Research Center of Engineering Technology for Utilization of Functional Ingredients from Botanicals, College of Horticulture, Hunan Agricultural University, Changsha 410128, China; (S.X.); (Y.Z.); (X.H.); (J.H.); (K.W.)
- Hunan Provincial Key Laboratory for Germplasm Innovation and Utilization of Crop, Hunan Agricultural University, Changsha 410128, China
| | - Kunbo Wang
- Key Laboratory of Tea Science of Ministry of Education, National Research Center of Engineering Technology for Utilization of Functional Ingredients from Botanicals, College of Horticulture, Hunan Agricultural University, Changsha 410128, China; (S.X.); (Y.Z.); (X.H.); (J.H.); (K.W.)
- Hunan Provincial Key Laboratory for Germplasm Innovation and Utilization of Crop, Hunan Agricultural University, Changsha 410128, China
| | - Zhonghua Liu
- Key Laboratory of Tea Science of Ministry of Education, National Research Center of Engineering Technology for Utilization of Functional Ingredients from Botanicals, College of Horticulture, Hunan Agricultural University, Changsha 410128, China; (S.X.); (Y.Z.); (X.H.); (J.H.); (K.W.)
- Hunan Provincial Key Laboratory for Germplasm Innovation and Utilization of Crop, Hunan Agricultural University, Changsha 410128, China
- Correspondence: (L.Y.); (Z.L.)
| |
Collapse
|
8
|
Hwang J, Newton EM, Hsiao J, Shi VY. Aryl Hydrocarbon Receptor/nuclear factor E2-related factor 2 (AHR/NRF2) Signaling: A Novel Therapeutic Target for Atopic Dermatitis. Exp Dermatol 2022; 31:485-497. [PMID: 35174548 DOI: 10.1111/exd.14541] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 01/25/2022] [Accepted: 02/12/2022] [Indexed: 11/28/2022]
Abstract
Aryl hydrocarbon receptor (AHR)/nuclear factor-erythroid 2-related factor 2 (NRF2) modulation are emerging as novel targets in the treatment of atopic dermatitis and other inflammatory skin disorders. Agonist activation of this pathway has downstream effects on epidermal barrier function, immunomodulation, oxidative stress reduction, and cutaneous microbiome modulation. Tapinarof, a dual agonist of the AHR/NRF2 signaling pathway, has shown promise in phase 2 trials for atopic dermatitis. In this review, we summarize current knowledge of the AHR/NRF2 pathway and implications in skin disease process. We also review the therapeutic potential of current AHR agonists and propose future directions to address knowledge gaps.
Collapse
Affiliation(s)
- Jonwei Hwang
- University of Illinois College of Medicine, 808 S. Wood St. - 380 CME, Chicago, IL, 60612-7307, USA
| | - Edita M Newton
- University of Arkansas for Medical Sciences, Department of Dermatology, 4301 West Markham, Slot 576, Little Rock, Arkansas, 72205, USA
| | - Jennifer Hsiao
- University of Southern California, Department of Dermatology, Ezralow Tower, 1441 Eastlake Avenue, Suite 5301, Los Angeles, CA, 90033, USA
| | - Vivian Y Shi
- University of Arkansas for Medical Sciences, Department of Dermatology, 4301 West Markham, Slot 576, Little Rock, Arkansas, 72205, USA
| |
Collapse
|
9
|
Sacran polysaccharide improves atopic dermatitis through inhibiting Th2 type immune response. Life Sci 2022; 288:120205. [PMID: 34871665 DOI: 10.1016/j.lfs.2021.120205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/30/2021] [Accepted: 11/30/2021] [Indexed: 12/20/2022]
Abstract
AIMS This study was aimed to explore whether sacran polysaccharide has a therapeutic effect on atopic dermatitis (AD) and its possible mechanisms. MATERIALS AND METHODS 2, 4-dinitrochlorobenzene (DNCB)-induced AD mice were treated with 0.2% Sacran, 0.5% Sacran and 0.1% tacrolimus. Through scoring dermatitis severity, measuring ear thickness, cracking behavior, open field test, we evaluated the therapeutic effect of Sacran on DNCB-induced AD mice. CD4+ T cells and CD8+ T cells were evaluated by flow cytometry. The relative expression of Ifng and Il4 were measured by real-time quantitative PCR. KEY FINDINGS Sacran could relieved the symptoms of DNCB-induced AD mice, such as AD score, ear thickness, and IgE release. Sacran may alleviate dermatitis by inhibiting Th2 activation and reducing IgE release. SIGNIFICANCE Our research further proved that polysaccharide Sacran has anti-dermatitis effects, and also clarified its mechanism of alleviating dermatitis by inhibiting the activation of Th2 cells and reducing the release of IgE, which provides a theoretical basis for the future clinical transformation of polysaccharide Sacran.
Collapse
|
10
|
Kishimoto R, Kato N, Koike M, Iwashita N, Takagi Y, Fukuyama T. Topical treatment with mastic (resin from Pistacia lentiscus) elicits anti-inflammatory and anti-pruritic responses by modulating keratinocyte activation in a mouse model of allergic dermatitis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 91:153679. [PMID: 34332283 DOI: 10.1016/j.phymed.2021.153679] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 07/01/2021] [Accepted: 07/15/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND As the number of patients with skin allergies, including atopic dermatitis, has increased rapidly, therapeutic options such as anti-IL-31 antibody and Janus kinase inhibitor have been developed recently. However, many concerns remain regarding the adverse effects and cost of these drugs; therefore, development of supplements that could support the effect of therapeutic agents is always required. PURPOSE The aim of this study was to develop preventive and supportive options for skin allergies by focusing on a natural product called "Mastic". METHODS Initially, the anti-inflammatory and anti-pruritic responses of 3% and 30% Mastic topical treatment were investigated in a mouse model of allergic contact dermatitis, generated by topical application of toluene-2,4-diisocyanate (TDI), a hapten that induces type 2 helper T cells. After itch behaviour and ear-swelling response were monitored, serum, auricular lymph nodes, and skin tissues were collected to analyse immunocyte differentiation, cytokine determination, and histological changes. RESULTS Our findings indicated that topical treatment with mastic significantly ameliorated ear swelling, itch behaviour, immunocyte infiltration, and cytokine production. Histological evaluation confirmed the occurrence of anti-inflammatory responses. The anti-inflammatory and anti-pruritic effects of topical treatment with mastic (3% and 5%) were further confirmed in a mouse model of atopic dermatitis which was generated by topical application of TDI in NC/Nga mice. Thickness of the back skin, AD score, transepidermal water loss (TEWL), and itch behaviour were measured weekly, and immunocyte differentiation, cytokine determination, and histological changes were also analysed. Mastic treatment significantly attenuated the skin thickness, AD score, TEWL, and itch behaviour. Corroborated reduction was observed in the numbers of T cells and IgE-B cells, as well as in pro-inflammatory cytokine production. The reproducibility of the effects of mastic was confirmed with 1% mastic ointment in a setting similar to the AD mouse model. In vitro evaluation of keratinocytes indicated that mastic pre-exposure induced a significant dose-dependent decrease in cytokine production. CONCLUSION Our findings thus demonstrate that topical treatment with mastic significantly ameliorate inflammatory and pruritic responses in a mouse model of allergic dermatitis.
Collapse
Affiliation(s)
- Risako Kishimoto
- Laboratory of Veterinary Pharmacology, Azabu University, 1-17-71, Fuchinobe, Chuo-ku, Sagamihara-shi, Kanagawa, Japan
| | - Narumi Kato
- Laboratory of Veterinary Pharmacology, Azabu University, 1-17-71, Fuchinobe, Chuo-ku, Sagamihara-shi, Kanagawa, Japan
| | - Mayuka Koike
- Laboratory of Veterinary Pharmacology, Azabu University, 1-17-71, Fuchinobe, Chuo-ku, Sagamihara-shi, Kanagawa, Japan
| | - Naoki Iwashita
- Laboratory of Veterinary Pharmacology, Azabu University, 1-17-71, Fuchinobe, Chuo-ku, Sagamihara-shi, Kanagawa, Japan; Bioalchemis, 3-28-61 Honshuku-cho, Fuchu-shi, Tokyo, Japan
| | - Yoshiichi Takagi
- Laboratory of Veterinary Pharmacology, Azabu University, 1-17-71, Fuchinobe, Chuo-ku, Sagamihara-shi, Kanagawa, Japan; Japan SLC, Inc, 85 Ohara-cho, Kita-ku, Hamamatsu-shi, Shizuoka, Japan
| | - Tomoki Fukuyama
- Laboratory of Veterinary Pharmacology, Azabu University, 1-17-71, Fuchinobe, Chuo-ku, Sagamihara-shi, Kanagawa, Japan.
| |
Collapse
|
11
|
Segner H, Bailey C, Tafalla C, Bo J. Immunotoxicity of Xenobiotics in Fish: A Role for the Aryl Hydrocarbon Receptor (AhR)? Int J Mol Sci 2021; 22:ijms22179460. [PMID: 34502366 PMCID: PMC8430475 DOI: 10.3390/ijms22179460] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 02/07/2023] Open
Abstract
The impact of anthropogenic contaminants on the immune system of fishes is an issue of growing concern. An important xenobiotic receptor that mediates effects of chemicals, such as halogenated aromatic hydrocarbons (HAHs) and polyaromatic hydrocarbons (PAHs), is the aryl hydrocarbon receptor (AhR). Fish toxicological research has focused on the role of this receptor in xenobiotic biotransformation as well as in causing developmental, cardiac, and reproductive toxicity. However, biomedical research has unraveled an important physiological role of the AhR in the immune system, what suggests that this receptor could be involved in immunotoxic effects of environmental contaminants. The aims of the present review are to critically discuss the available knowledge on (i) the expression and possible function of the AhR in the immune systems of teleost fishes; and (ii) the impact of AhR-activating xenobiotics on the immune systems of fish at the levels of immune gene expression, immune cell proliferation and immune cell function, immune pathology, and resistance to infectious disease. The existing information indicates that the AhR is expressed in the fish immune system, but currently, we have little understanding of its physiological role. Exposure to AhR-activating contaminants results in the modulation of numerous immune structural and functional parameters of fish. Despite the diversity of fish species studied and the experimental conditions investigated, the published findings rather uniformly point to immunosuppressive actions of xenobiotic AhR ligands in fish. These effects are often associated with increased disease susceptibility. The fact that fish populations from HAH- and PAH-contaminated environments suffer immune disturbances and elevated disease susceptibility highlights that the immunotoxic effects of AhR-activating xenobiotics bear environmental relevance.
Collapse
Affiliation(s)
- Helmut Segner
- Centre for Fish and Wildlife Health, Department of Pathobiology and Infectious Diseases, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| | | | | | - Jun Bo
- Laboratory of Marine Biology and Ecology, Third Institute of Oceanography, Xiamen 361005, China
| |
Collapse
|
12
|
Yanagisawa R, Koike E, Takano H. Benzo[a]pyrene aggravates atopic dermatitis-like skin lesions in mice. Hum Exp Toxicol 2021; 40:S269-S277. [PMID: 34424081 DOI: 10.1177/09603271211036123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background: Benzo[a]pyrene (BaP) affects the immune system and causes mutagenic and carcinogenic effects. Purpose: We aimed to evaluate the effects of systemic exposure to BaP on mite allergen-induced atopic dermatitis (AD)-like skin lesions in mice. Methods: Mite allergen (Dermatophagoides pteronyssinus; Dp) was injected intradermally into the right ears of NC/Nga male mice on eight occasions every 2-3 days. Benzo[a]pyrene was administered intraperitoneally in the equivalent doses of 0, 2, 20, 200, or 2000 μg/kg/day, once a week on four occasions. Results: AD-like skin inflammation related to mite allergen worsened by BaP exposure at 2, 20 µg/kg/day doses; this was in parallel with eosinophil and mast cell infiltration and mast cell degranulation. A trend was also observed toward increased proinflammatory molecule expression, including macrophage inflammatory protein-1 alpha, interleukin (IL)-4, IL-13, and IL-18, in the ear tissue. However, 200 or 2000 µg/kg/day BaP attenuated the enhancing effects. In the regional lymph nodes, 2 µg/kg/day BaP with Dp enhanced antigen-presenting cell and T cell activation compared with Dp alone. Conclusions: This suggests that BaP exposure can aggravate Dp-induced AD-like skin lesions through TH2-biased responses in the inflamed sites and the activation of regional lymph nodes. Therefore, BaP may be responsible for the recent increase in AD incidence.
Collapse
Affiliation(s)
- Rie Yanagisawa
- Health and Environmental Risk Division, 13585National Institute for Environmental Studies, Tsukuba, Japan
| | - Eiko Koike
- Health and Environmental Risk Division, 13585National Institute for Environmental Studies, Tsukuba, Japan
| | - Hirohisa Takano
- Graduate School of Global Environmental Studies, 12918Kyoto University, Kyoto, Japan
| |
Collapse
|
13
|
Anti-Inflammatory Activity of a Medicinal Herb Extract Mixture, HM-V, on an Animal Model of DNCB-Induced Chronic Skin Inflammation. PLANTS 2021; 10:plants10081546. [PMID: 34451592 PMCID: PMC8400547 DOI: 10.3390/plants10081546] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/26/2021] [Accepted: 07/26/2021] [Indexed: 11/25/2022]
Abstract
Chronic inflammatory skin diseases, such as atopic dermatitis, are caused by the accumulation of immune cells and the overproduction of chemokines, including CCL17 and CCL22, due to the activation of pro-inflammatory cytokines secreted from keratinocytes. In the present study, the inhibitory activity of HM-V on tumor necrosis factor alpha (TNF-α)/interferon gamma (IFN-γ)-induced pro-inflammatory cytokines was examined in human keratinocytes (HaCaTs) and 2,4-dinitrofluorobenzene (DNCB)-induced chronic skin contact dermatitis animal models. Traditional Asian medicinal herb extracts mixture (HM-V), which have been extensively used in Asian medicine, were utilized. In TNF-α/IFN-γ-induced HaCaTs, HM-V strongly inhibited mRNA and protein expression of CCL17 and CCL22 in a concentration-dependent manner. The expression of pro-inflammatory cytokines such as TNF-α, IL-1β, and IL-6 was also inhibited. Therefore, localized administration of HM-V in the DNCB-induced animal model alleviated immune cell deposition and skin inflammation. The results indicate that HM-V exerts inhibitory effects on keratinocyte production of CCL17 and CCL22. Furthermore, HM-V may be a useful anti-inflammatory agent for the prevention and treatment of inflammatory skin diseases.
Collapse
|
14
|
Wang E, Tu W, Do DC, Xiao X, Bhatti SB, Yang L, Sun X, Xu D, Yang P, Huang SK, Gao P, Liu Z. Benzo(a)pyrene Enhanced Dermatophagoides Group 1 (Der f 1)-Induced TGFβ1 Signaling Activation Through the Aryl Hydrocarbon Receptor-RhoA Axis in Asthma. Front Immunol 2021; 12:643260. [PMID: 33936062 PMCID: PMC8081905 DOI: 10.3389/fimmu.2021.643260] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 02/24/2021] [Indexed: 12/18/2022] Open
Abstract
We have previously demonstrated that benzo(a)pyrene (BaP) co-exposure with dermatophagoides group 1 allergen (Der f 1) can potentiate Der f 1-induced airway inflammation. The underlying mechanism, however, remains undetermined. Here we investigated the molecular mechanisms underlying the potentiation of BaP exposure on Der f 1-induced airway inflammation in asthma. We found that BaP co-exposure potentiated Der f 1-induced TGFβ1 secretion and signaling activation in human bronchial epithelial cells (HBECs) and the airways of asthma mouse model. Moreover, BaP exposure alone or co-exposure with Der f 1-induced aryl hydrocarbon receptor (AhR) activity was determined by using an AhR-dioxin-responsive element reporter plasmid. The BaP and Der f 1 co-exposure-induced TGFβ1 expression and signaling activation were attenuated by either AhR antagonist CH223191 or AhR knockdown in HBECs. Furthermore, AhR knockdown led to the reduction of BaP and Der f 1 co-exposure-induced active RhoA. Inhibition of RhoA signaling with fasudil, a RhoA/ROCK inhibitor, suppressed BaP and Der f 1 co-exposure-induced TGFβ1 expression and signaling activation. This was further confirmed in HBECs expressing constitutively active RhoA (RhoA-L63) or dominant-negative RhoA (RhoA-N19). Luciferase reporter assays showed prominently increased promoter activities for the AhR binding sites in the promoter region of RhoA. Inhibition of RhoA suppressed BaP and Der f 1 co-exposure-induced airway hyper-responsiveness, Th2-associated airway inflammation, and TGFβ1 signaling activation in asthma. Our studies reveal a previously unidentified functional axis of AhR-RhoA in regulating TGFβ1 expression and signaling activation, representing a potential therapeutic target for allergic asthma.
Collapse
Affiliation(s)
- Eryi Wang
- Department of Respiratory and Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
- The State Key Laboratory of Respiratory Disease for Allergy, Shenzhen Key Laboratory of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Wei Tu
- Department of Respiratory and Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
- The State Key Laboratory of Respiratory Disease for Allergy, Shenzhen Key Laboratory of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
- Johns Hopkins Asthma and Allergy Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Danh C. Do
- Johns Hopkins Asthma and Allergy Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Xiaojun Xiao
- The State Key Laboratory of Respiratory Disease for Allergy, Shenzhen Key Laboratory of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Shehar B. Bhatti
- Johns Hopkins Asthma and Allergy Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Liteng Yang
- Department of Respiratory and Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Xizhuo Sun
- Department of Respiratory and Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Damo Xu
- Department of Respiratory and Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
- The State Key Laboratory of Respiratory Disease for Allergy, Shenzhen Key Laboratory of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Pingchang Yang
- Department of Respiratory and Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
- The State Key Laboratory of Respiratory Disease for Allergy, Shenzhen Key Laboratory of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Shau-Ku Huang
- Department of Respiratory and Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
- Johns Hopkins Asthma and Allergy Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli, Taiwan
| | - Peisong Gao
- Johns Hopkins Asthma and Allergy Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Zhigang Liu
- Department of Respiratory and Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
- The State Key Laboratory of Respiratory Disease for Allergy, Shenzhen Key Laboratory of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
| |
Collapse
|
15
|
Ookawara T, Aihara R, Morimoto A, Iwashita N, Kurata K, Takagi Y, Miyasaka A, Kushiro M, Miyake S, Fukuyama T. Acute and subacute oral toxicity of deoxynivalenol exposure in a Dermatophagoides farinae induced murine asthma model. Toxicol Sci 2020; 179:229-240. [PMID: 33170220 DOI: 10.1093/toxsci/kfaa168] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Previously, researchers have demonstrated that mycotoxin deoxynivalenol significantly enhances immunocyte activation. However, the interaction between deoxynivalenol exposure and immune disorders remains unclear. In this study, we aimed to investigate whether acute and subacute oral exposure to deoxynivalenol exacerbates the development of respiratory allergy using a mite allergen (Dermatophagoides farina, Derf)-induced mouse model of asthma. The direct relationship between deoxynivalenol exposure and asthma development was examined following acute oral deoxynivalenol administration (0, 0.1, or 0.3 mg/kg body weight), immediately before the final mite allergen challenge. Simultaneously, the influence of subacute oral exposure via low dose deoxynivalenol contaminated wheat (0.33 ppm) was evaluated using the same settings. To detect the pro-inflammatory effects of deoxynivalenol exposure, we examined the total and Derf-specific serum IgE levels, histology, number of immunocytes, and cytokine and chemokine secretion. Acute oral deoxynivalenol significantly enhanced the inflammatory responses, including cellular infiltration into bronchoalveolar lavage fluid, infiltration of immunocytes and cytokine production in local lymph nodes, and cytokine levels in lung tissues. Corresponding pro-inflammatory responses were observed in a mouse group exposed to subacute oral deoxynivalenol. In vivo results were validated by in vitro experiments using the human bronchial epithelial (BEAS-2B) and human eosinophilic leukemia (EOL-1) cell lines. Following exposure to deoxynivalenol, the secretion of interleukin (IL)-1β, IL-6, IL-8, and/or tumor necrosis factor (TNF)-α in BEAS-2B cells, as well as EoL-1 cells, increased significantly. Our findings indicate that deoxynivalenol exposure is significantly involved in the pro-inflammatory response observed in respiratory allergy.
Collapse
Affiliation(s)
- Toa Ookawara
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara-shi, Kanagawa, Japan
| | - Ryota Aihara
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara-shi, Kanagawa, Japan
| | - Ai Morimoto
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara-shi, Kanagawa, Japan
| | - Naoki Iwashita
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara-shi, Kanagawa, Japan.,Bioalchemis, 3-28-61 Honshuku-cho, Fuchu-shi, Tokyo, Japan
| | - Keigo Kurata
- Institute of Tokyo Environmental Allergy, ITEA Inc, 1-2-5, Yushima, Bunkyo-ku, Tokyo, Japan
| | - Yoshiichi Takagi
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara-shi, Kanagawa, Japan.,Japan SLC, Inc, 85 Ohara-cho, Kita-ku, Hamamatsu-shi, Shizuoka, Japan
| | - Atsushi Miyasaka
- Kyushu Okinawa Agricultural Research Center, National Agriculture and Food Research Organization (NARO), Suya 2421, Koshi, Kumamoto, Japan
| | - Masayo Kushiro
- Food Research Institute, National Agriculture and Food Research Organization (NARO), 2-1-12 Kannondai, Tsukuba, Ibaraki, Japan
| | - Shiro Miyake
- Laboratory of Food and Hygiene, School of Food and Life Science, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara-shi, Kanagawa, Japan
| | - Tomoki Fukuyama
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara-shi, Kanagawa, Japan
| |
Collapse
|
16
|
Wang F, Liang S, Hu J, Xu Y. Aryl hydrocarbon receptor connects dysregulated immune cells to atherosclerosis. Immunol Lett 2020; 228:55-63. [PMID: 33053378 DOI: 10.1016/j.imlet.2020.10.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/29/2020] [Accepted: 10/08/2020] [Indexed: 11/20/2022]
Abstract
As a chronic inflammatory disease with autoimmune components, atherosclerosis is the major cause of cardiovascular morbidity and mortality. Recent studies have revealed that the development of atherosclerosis is strongly linked to the functional activities of aryl hydrocarbon receptor (AHR), a chemical sensor that is also important for the development, maintenance, and function of a variety of immune cells. In this review, we focus on the impact of AHR signaling on the different cell types that are closely related to the atherogenesis, including T cells, B cells, dendritic cells, macrophages, foam cells, and hematopoietic stem cells in the arterial walls, and summarize the latest development on the interplay between this environmental sensor and immune cells in the context of atherosclerosis. Hopefully, elucidation of the role of AHR in atherosclerosis will facilitate the understanding of case variation in disease prevalence and may aid in the development of novel therapies.
Collapse
Affiliation(s)
- Fengge Wang
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, School of Life Science, Anhui Normal University, Wuhu, 241000, China
| | - Shuangchao Liang
- Department of Vascular Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, 241000, China
| | - Jiqiong Hu
- Department of Vascular Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, 241000, China
| | - Yuekang Xu
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, School of Life Science, Anhui Normal University, Wuhu, 241000, China.
| |
Collapse
|
17
|
Aihara R, Ookawara T, Morimoto A, Iwashita N, Takagi Y, Miyasaka A, Kushiro M, Miyake S, Fukuyama T. Acute and subacute oral administration of mycotoxin deoxynivalenol exacerbates the pro-inflammatory and pro-pruritic responses in a mouse model of allergic dermatitis. Arch Toxicol 2020; 94:4197-4207. [DOI: 10.1007/s00204-020-02875-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 08/12/2020] [Indexed: 12/24/2022]
|
18
|
Li N, Gao S, Tong J, Yu Y, Zhang Q, Xu C. Probiotics as a functional food ingredient in allergic diseases: regulation of CD4+ T helper cell differentiation. Crit Rev Microbiol 2020; 46:463-474. [PMID: 32720543 DOI: 10.1080/1040841x.2020.1796578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 10/23/2022]
Abstract
Allergic diseases are increasing worldwide, associating with increased health costs and decreased quality of life. Allergy is immune-related diseases caused by an allergic immune response to innocuous substance in the environment. At present, research has focussed on the study of the relevance to the microbiome and the phenotypes of allergy, including the relationships among the gastrointestinal microbiome, immune function, and allergic sensitisation. Probiotics as functional food ingredient are thought to secrete functional metabolites that have antibacterial effects on ameliorating intestinal health and CD4+ T helper cells-mediated immunity. This review will summarise the role of probiotics in the immune regulation and flora balance, highlighting recent advances in our understanding of the imbalance of Th subsets and cytokine leading to the immunopathology of allergic reactions. Finally, we discussed the unresolved problems and future research directions in order to promote the clinical application of probiotics immunotherapy.
Collapse
Affiliation(s)
- Na Li
- Pediatric Department, School of Medicine, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, China
- Institute of Tropical Medicine, Hainan Medical University, HaiKou, China
| | - Shenshen Gao
- Pediatric Department, School of Medicine, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jie Tong
- College of Life Science, Institute of Life Science and Green Development, Hebei University, Baoding, China
| | - Yi Yu
- Pediatric Department, School of Medicine, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Qingqing Zhang
- Pediatric Department, School of Medicine, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Chundi Xu
- Pediatric Department, School of Medicine, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
19
|
Tajima H, Tajiki-Nishino R, Watanabe Y, Kurata K, Fukuyama T. Activation of aryl hydrocarbon receptor by benzo[a]pyrene increases interleukin 33 expression and eosinophil infiltration in a mouse model of allergic airway inflammation. J Appl Toxicol 2020; 40:1545-1553. [PMID: 32557721 DOI: 10.1002/jat.4017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 05/05/2020] [Accepted: 05/16/2020] [Indexed: 12/15/2022]
Abstract
We recently demonstrated that benzo[a]pyrene (BaP), the aryl hydrocarbon receptor (AhR) ligand, directly contributes to aggravation of cutaneous allergy in a mouse model of allergic dermatitis. The present study aimed to determine whether BaP-induced AhR activation results in development of airway inflammation. Initially, the potential for a direct relationship between BaP-induced AhR activation and airway inflammation was investigated in vivo, using a mouse model of type 2 helper T cell (Th2) hapten toluene-2,4-diisocyanate (TDI)-induced airway inflammation. Mice were orally administered BaP at 48, 24, and 4 h before the final allergen challenge. Oral administration of BaP showed a significant increase in lung inflammation and eosinophil infiltration. While expression of Th2 cytokines such as interleukin 4 (IL-4) and IL-13 was not affected by exposure to BaP, AhR activation significantly increased IL-33 expression. To confirm the in vivo results, in vitro experiments were performed using the human eosinophilic leukemia cell line (EOL-1), human bronchial epithelial cell line (BEAS-2B), and human lung adenocarcinoma epithelial cell line (A549). Results indicated that pre-treatment with BaP increased expression of IL-8 in house dust mite-activated EOL-1, BEAS-2B, and A549 cells. In addition, IL-33 levels in BEAS-2B cells were significantly increased after BaP exposure. Our findings indicated that BaP-induced AhR activation is involved in the pro-inflammatory response in respiratory allergy, and that this effect may be mediated by increased IL-33 expression and eosinophil infiltration.
Collapse
Affiliation(s)
- Hitoshi Tajima
- Toxicology Division, Institute of Environmental Toxicology, Ibaraki, Japan
| | | | - Yuko Watanabe
- Toxicology Division, Institute of Environmental Toxicology, Ibaraki, Japan
| | - Keigo Kurata
- Institute of Tokyo Environmental Allergy, ITEA Inc., Tokyo, Japan
| | - Tomoki Fukuyama
- Toxicology Division, Institute of Environmental Toxicology, Ibaraki, Japan.,Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Azabu University, Sagamihara-shi, Kanagawa, Japan
| |
Collapse
|