1
|
Zůvalová I, Vyhlídalová B, Ondrová K, Nádvorník P, Hrubý J, Illés P, Soural M, Šebela M, Šindlerová L, Kubala L, Mani S, Dvořák Z. Decoding structural determinants of aryl hydrocarbon receptor antagonism by monoterpenoids. Bioorg Chem 2025; 157:108265. [PMID: 39952059 DOI: 10.1016/j.bioorg.2025.108265] [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: 12/14/2024] [Revised: 02/08/2025] [Accepted: 02/08/2025] [Indexed: 02/17/2025]
Abstract
Monocyclic monoterpenoids carvones have been recently identified as atypical negative allosteric modulators of aryl hydrocarbon receptor (AhR). In the current work, we performed AhR antagonist activity screening of 100 natural and synthetic monoterpenoids, and their analogues. Using SAR approach, structural determinants of AhR antagonist activity were assigned, including CO presence/position, planarity, and C3/C5-alkylation. Applying pyramidal selection criteria, including absence of residual agonist activity, no cytotoxicity, strong antagonist potency, and pan-antagonism against diverse AhR agonists, we distilled four lead AhR antagonists (carvacrol, o-cresol, 3-methyl-S-carvone, EN-2). Whereas 3-methyl-S-carvone and EN-2 were non-competitive AhR pan-antagonists, carvacrol and o-cresol were ligand-selective AhR antagonists acting by unclear mechanism. We characterized in detail the effects of lead compounds at cellular functions of AhR, including AhR nuclear translocation, AhR dimerization with ARNT, and the expression of AhR-regulated genes. As a proof of concept, effects of monoterpenoids in the murine macrophages were investigated.
Collapse
Affiliation(s)
- Iveta Zůvalová
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Šlechtitelů 27, Olomouc 779 00, Czech Republic.
| | - Barbora Vyhlídalová
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Šlechtitelů 27, Olomouc 779 00, Czech Republic
| | - Karolína Ondrová
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Šlechtitelů 27, Olomouc 779 00, Czech Republic
| | - Petr Nádvorník
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Šlechtitelů 27, Olomouc 779 00, Czech Republic
| | - Jiří Hrubý
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Šlechtitelů 27, Olomouc 779 00, Czech Republic
| | - Peter Illés
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Šlechtitelů 27, Olomouc 779 00, Czech Republic
| | - Miroslav Soural
- Department of Organic Chemistry, Faculty of Science, Palacký University, 17. Listopadu 12, Olomouc 771 46, Czech Republic
| | - Marek Šebela
- Department of Biochemistry, Faculty of Science, Palacký University, Šlechtitelů 27, Olomouc 779 00, Czech Republic
| | - Lenka Šindlerová
- Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, Brno 612 00, Czech Republic
| | - Lukáš Kubala
- Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, Brno 612 00, Czech Republic
| | - Sridhar Mani
- Department of Medicine and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Zdeněk Dvořák
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Šlechtitelů 27, Olomouc 779 00, Czech Republic.
| |
Collapse
|
2
|
Banerjee O, Paul T, Singh S, Maji BK, Mukherjee S. Individual and combined antagonism of aryl hydrocarbon receptor (AhR) and estrogen receptors (ERs) offers distinct level of protection against Bisphenol A (BPA)-induced pancreatic islet cell toxicity in mice. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3939-3954. [PMID: 39377923 DOI: 10.1007/s00210-024-03506-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 09/30/2024] [Indexed: 10/09/2024]
Abstract
Bisphenol A (BPA), a pervasive endocrine-disrupting chemical, is known to convey harmful impact on pancreatic islets through estrogen receptors (ERs). Conversely, BPA can activate aryl hydrocarbon receptor (AhR) in certain contexts and has raised concerns about potential toxicological effects. However, BPA-AhR interaction in the context of pancreatic islet toxicity is yet to be reported. We demonstrated the specific role of AhR and its interaction with ERs to mediate BPA toxicity in pancreatic islets. In vitro, isolated islet cells treated with BPA (1 nM), with or without CH22319 (10 mM) and ICI182780 (1 mM) and insulin release, glucose-stimulated insulin secretion (GSIS), cell viability, and pERK1/2 and pAkt expression were measured. In vivo, mice were treated with BPA (10 and 100 µg/kg body weight/day for 21 days) with or without intraperitonial co-treatment of CH22319 (AhR antagonist, 10mg/kg), and ICI182780 (ER antagonist, 500 µg/kg). Glucose homeostasis, insulin resistance, oxidative stress, and inflammatory markers were measured. In vitro data revealed the involvement of AhR in the BPA-mediated alteration in insulin secretion, GSIS, and pERK1/2 and pAkt expression which were counteracted by CH223191 (AhR antagonist) alone or with ICI182780 (ER antagonist). Further, CH223191 alone or with ICI182780 modulated BPA-induced oxidative stress and pro-inflammatory cytokines and alleviated islet cell dysfunction and impaired insulin secretion. In conclusion, therapeutic targeting of AhR and ER combined might be a promising target against diabetogenic action of BPA.
Collapse
Affiliation(s)
- Oly Banerjee
- Department of Physiology, Serampore College, 9 William Carey Road, Serampore, Hooghly, 712201, West Bengal, India
- Department of Medical Laboratory Technology, School of Allied Health Sciences, Swami Vivekananda University, Bara Kanthalia, West Bengal, 700121, India
| | - Tiyesh Paul
- Department of Physiology, Serampore College, 9 William Carey Road, Serampore, Hooghly, 712201, West Bengal, India
| | - Siddhartha Singh
- Department of Physiology, Serampore College, 9 William Carey Road, Serampore, Hooghly, 712201, West Bengal, India
| | - Bithin Kumar Maji
- Department of Physiology, Serampore College, 9 William Carey Road, Serampore, Hooghly, 712201, West Bengal, India
| | - Sandip Mukherjee
- Department of Physiology, Serampore College, 9 William Carey Road, Serampore, Hooghly, 712201, West Bengal, India.
| |
Collapse
|
3
|
Qiu S, Ding X, Ma X, Zhang L, Chen J, Wei W. Muscle cells affect the promoting effect of FGF21 on lipid accumulation in porcine adipocytes through AhR/FGFR1 signaling pathway. Biochem Biophys Res Commun 2025; 754:151520. [PMID: 40015071 DOI: 10.1016/j.bbrc.2025.151520] [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: 02/12/2025] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/01/2025]
Abstract
The intramuscular fat (IMF) content, as an important meat quality trait, can directly affect the tenderness, juiciness, and flavor of pork. Reasonably increasing the IMF content can improve the palatability of pork. Therefore, identification of important factors for the lipid accumulation among muscles is the breakthrough point for improving meat quality. FGF21, identified as a novel metabolic regulator, has been found to regulate glucose and lipid metabolism in 3T3-L1 adipocytes, but its function in porcine adipocytes remains unclear. In this study, we discovered that the administration of recombinant FGF21 protein promotes adipogenic differentiation and increases triglyceride accumulation in porcine adipocytes. While the expression of FGFR1 in adipocytes under muscle conditions is inhibited, affecting the signal transduction of FGF21. This inhibitory effect is accompanied by activation of the AhR signaling pathway. When treated with the AhR antagonist CH223191, there was a partial restoration of FGFR1 expression levels. This indicates that muscle cells suppress the expression of FGFR1 in adipocytes by activating the AhR signaling pathway, thereby affecting the signal transduction of FGF21. Our results reveal the regulatory role of FGF21 in pig adipocyte differentiation and the regulatory mechanism of muscle environment on FGFR1 expression, providing new theoretical basis for IMF content improvement from the perspective of FGF21-FGFR1 signaling transduction.
Collapse
Affiliation(s)
- Shengda Qiu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Xiaolei Ding
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Xiangfei Ma
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Lifan Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Jie Chen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Wei Wei
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
4
|
Wang Y, Quan L, Zheng X, Hu Q, Huang X, Pu Y, Xie G, Peng Q. Indole compounds from fermented soybean products activate the aryl hydrocarbon receptor to reduce liver injury. NPJ Sci Food 2025; 9:38. [PMID: 40122901 PMCID: PMC11930980 DOI: 10.1038/s41538-025-00404-z] [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: 07/11/2024] [Accepted: 03/03/2025] [Indexed: 03/25/2025] Open
Abstract
The consumption of stinky tofu, a traditional fermented soybean product from China, elevates the concentrations of indole and trimethylindole in murine feces and increases the levels of indole in serum, as well as indole in the liver. These hepatic compounds act as ligands for the Aryl Hydrocarbon Receptor (AHR), triggering activation of this receptor, which subsequently enhances the expression of the enzyme cytochrome P450 (CYP) 1A1. This upregulation diminishes the levels of pro-inflammatory cytokines, thereby attenuating alcohol-induced liver injury. This study underscores the potential of dietary indole from stinky tofu to mitigate Alcoholic liver disease (ALD), laying a foundation for the development of functional foods and novel treatment strategies for ALD.
Collapse
Affiliation(s)
- Yanyun Wang
- College of Life Science, Leshan Normal University, Leshan, China
| | - Leping Quan
- National Engineering Research Center for Chinese CRW (branch center), School of Life and Environmental Sciences, Shaoxing University, Shaoxing, China
| | - Xiaomin Zheng
- Wuxi Maternity and Child Health Care Hospital, Affiliated Women's Hospital of Jiangnan University, Wuxi, China
| | - Qiang Hu
- College of Life Science, Leshan Normal University, Leshan, China
| | - Xiaoli Huang
- Shaoxing Testing Institute of Quality and Technical Supervision, Shaoxing, China
| | - Yang Pu
- Shaoxing Testing Institute of Quality and Technical Supervision, Shaoxing, China
| | - Guangfa Xie
- Zhejiang Collaborative Innovation Center for Full-Process Monitoring and Green Governance of Emerging Contaminants, College of Biology and Environmental Engineering, Zhejiang Shuren University, Hangzhou, China.
| | - Qi Peng
- National Engineering Research Center for Chinese CRW (branch center), School of Life and Environmental Sciences, Shaoxing University, Shaoxing, China.
| |
Collapse
|
5
|
Del Sorbo L, Acconcia C, Salvatore MM, Fusco G, Vasinioti V, Lucente MS, Zhu L, Pratelli A, Russo L, Andolfi A, Iacovino R, Fiorito F. Insight into the Role of the Aryl Hydrocarbon Receptor in Bovine Coronavirus Infection by an Integrated Approach Combining In Vitro and In Silico Methods. Microorganisms 2025; 13:579. [PMID: 40142473 PMCID: PMC11944835 DOI: 10.3390/microorganisms13030579] [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: 01/23/2025] [Revised: 02/19/2025] [Accepted: 02/26/2025] [Indexed: 03/28/2025] Open
Abstract
It is well known that the host response to different human and animal coronaviruses infection is regulated by the aryl hydrocarbon receptor, a ligand-activated transcription factor. The present study investigates the expression of the aryl hydrocarbon receptor during bovine coronavirus infection, through in vitro and in silico investigations. The in vitro studies demonstrate that the aryl hydrocarbon receptor and as well as its targets, CYP1A1 and CYP1B1, were significantly activated by bovine coronavirus infection in bovine cells (MDBK). During infection, the pretreatment of cells with non-cytotoxic doses of CH223191, a selective inhibitor of the aryl hydrocarbon receptor, resulted in a significant reduction in virus yield and a downregulation in the viral spike protein expression. These findings occurred in the presence of the inhibition of aryl hydrocarbon receptor signaling. Our results reveal that the bovine coronavirus acts on viral replication, upregulating the aryl hydrocarbon receptor and its downstream target proteins, CYP1A1 and CYP1B1. In addition, following the in silico studies, the three-dimensional structural model of the bovine aryl hydrocarbon receptor in complex with the antagonist CH223191 indicates that the molecular mechanism, by which the PASB and TAD domains of the receptor interact with the inhibitor, is mainly driven by an extensive network of hydrophobic interactions, with a series of hydrogen bonds contributing to stabilizing the complex. Interestingly, bioinformatic analyses revealed that the PASB and TAD domains in the human and bovine aryl hydrocarbon receptor present high similarity at the primary sequence and three-dimensional structure levels. Taken together, these findings represent a fundamental step for the development of innovative drugs targeting AhR as a potential object for CoVs therapy.
Collapse
Affiliation(s)
- Luca Del Sorbo
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy; (L.D.S.); (M.M.S.)
| | - Clementina Acconcia
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, 81100 Caserta, Italy; (C.A.); (L.R.)
| | - Maria Michela Salvatore
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy; (L.D.S.); (M.M.S.)
| | - Giovanna Fusco
- Istituto Zooprofilattico Sperimentale del Mezzogiorno, 80055 Portici, Italy
| | - Violetta Vasinioti
- Department of Veterinary Medicine, University of Bari, 70010 Valenzano, Italy; (V.V.); (M.S.L.); (A.P.)
| | - Maria Stella Lucente
- Department of Veterinary Medicine, University of Bari, 70010 Valenzano, Italy; (V.V.); (M.S.L.); (A.P.)
| | - Liqian Zhu
- College of Life Sciences, Hebei University, Baoding 071002, China;
| | - Annamaria Pratelli
- Department of Veterinary Medicine, University of Bari, 70010 Valenzano, Italy; (V.V.); (M.S.L.); (A.P.)
| | - Luigi Russo
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, 81100 Caserta, Italy; (C.A.); (L.R.)
| | - Anna Andolfi
- Department of Chemical Science, University of Naples Federico II, 80126 Naples, Italy;
| | - Rosa Iacovino
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, 81100 Caserta, Italy; (C.A.); (L.R.)
| | - Filomena Fiorito
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy; (L.D.S.); (M.M.S.)
| |
Collapse
|
6
|
Barreira-Silva P, Lian Y, Kaufmann SHE, Moura-Alves P. The role of the AHR in host-pathogen interactions. Nat Rev Immunol 2025; 25:178-194. [PMID: 39415055 DOI: 10.1038/s41577-024-01088-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2024] [Indexed: 10/18/2024]
Abstract
Host-microorganism encounters take place in many different ways and with different types of outcomes. Three major types of microorganisms need to be distinguished: (1) pathogens that cause harm to the host and must be controlled; (2) environmental microorganisms that can be ignored but must be controlled at higher abundance; and (3) symbiotic microbiota that require support by the host. Recent evidence indicates that the aryl hydrocarbon receptor (AHR) senses and initiates signalling and gene expression in response to a plethora of microorganisms and infectious conditions. It was originally identified as a receptor that binds xenobiotics. However, it was subsequently found to have a critical role in numerous biological processes, including immunity and inflammation and was recently classified as a pattern recognition receptor. Here we review the role of the AHR in host-pathogen interactions, focusing on AHR sensing of different microbial classes, the ligands involved, responses elicited and disease outcomes. Moreover, we explore the therapeutic potential of targeting the AHR in the context of infection.
Collapse
Affiliation(s)
- Palmira Barreira-Silva
- IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Yilong Lian
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Stefan H E Kaufmann
- Max Planck Institute for Infection Biology, Berlin, Germany
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Hagler Institute for Advanced Study, Texas A&M University, College Station, TX, USA
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Pedro Moura-Alves
- IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
7
|
Wei KL, Chen SC, Lin CY, Chou YT, Kuo WT, Chuah TW, Joseph Su JG. Dexlansoprazole is an aryl hydrocarbon receptor agonist. Food Chem Toxicol 2025; 197:115262. [PMID: 39832710 DOI: 10.1016/j.fct.2025.115262] [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: 10/01/2024] [Revised: 12/14/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
Dexlansoprazole, a proton pump inhibitor, is commonly used to treat gastro-esophageal reflux disease and erosive esophagitis. The activated aryl hydrocarbon receptor (AhR) functions as a transcription factor by binding to the aryl hydrocarbon response element (AHRE) of its target genes, with cytochrome P450 (CYP) 1A1 being the most well-known target. In this study, we demonstrated that dexlansoprazole stimulates AhR activity, leading to increased CYP1A1 expression. Our findings indicate that treatment with 2 μM dexlansoprazole is sufficient to induce CYP1A1 mRNA and protein expression, as well as AHRE-mediated transcriptional activity, in both human and mouse cells. Using AhR signal-deficient mutant cells and specific AhR antagonists-SR1, GNF351, and CH-223191-we confirmed that AhR is required for dexlansoprazole-induced CYP1A1 expression. Additionally, we showed that dexlansoprazole promotes AhR nuclear translocation, acting as an AhR agonist. However, due to its lower potency compared to FICZ and ITE in activating AhR, dexlansoprazole suppresses FICZ- and ITE-induced CYP1A1 expression in human liver HepG2 and ovarian granulosa HO23 cell lines, suggesting that it functions as both an AhR agonist and a modulator. This study offers valuable insights into the potential clinical side effects of dexlansoprazole.
Collapse
Affiliation(s)
- Kuo-Liang Wei
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chiayi, 61363, Taiwan, ROC; College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan, ROC
| | - Shan-Chun Chen
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi, 60004, Taiwan, ROC
| | - Chih-Yi Lin
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi, 60004, Taiwan, ROC
| | - Yu-Ting Chou
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi, 60004, Taiwan, ROC
| | - Wei-Tin Kuo
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi, 60004, Taiwan, ROC
| | - Teik-Wei Chuah
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi, 60004, Taiwan, ROC
| | - Jyan-Gwo Joseph Su
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi, 60004, Taiwan, ROC.
| |
Collapse
|
8
|
Wuputra K, Hsu WH, Ku CC, Yang YH, Kuo KK, Yu FJ, Yu HS, Nagata K, Wu DC, Kuo CH, Yokoyama KK. The AHR-NRF2-JDP2 gene battery: Ligand-induced AHR transcriptional activation. Biochem Pharmacol 2025; 233:116761. [PMID: 39855429 DOI: 10.1016/j.bcp.2025.116761] [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: 11/02/2024] [Revised: 12/18/2024] [Accepted: 01/16/2025] [Indexed: 01/27/2025]
Abstract
Aryl hydrocarbon receptor (AHR) and nuclear factor-erythroid 2-related factor 2 (NRF2) can regulate a series of genes encoding the detoxifying phase I and II enzymes, via a signaling crosstalk known as the "AHR-NRF2 gene battery". The chromatin transcriptional regulator Jun dimerization protein 2 (JDP2) plays a central role in thetranscription of AHR gene in response to the phase I enzyme ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin. It forms a transcriptional complex with AHR-AHR nuclear translocator (ARNT) and NRF2-small musculoaponeurotic fibrosarcoma proteins (sMAF), which are then recruited to the respective cis-elements, such as dioxin response elements and antioxidant response elements, respectively, in the AHR promoter. Here, we present a revised description of the AHR-NRF2 gene battery as the AHR-NRF2-JDP2 gene battery for transactivating the AHR promoter by phase I enzyme ligands. The chromatin regulator JDP2 was found to be involved in the movement of AHR-NRF2 complexes from the dioxin response element to the antioxidant response element in the AHR promoter, during its activation in a spatiotemporal manner. This new epigenetic and chromatin remodeling role of AHR-NRF2-JDP2 axis is useful for identifying new therapeutic targets for various diseases, including immunological response, detoxification, development, and cancer-related diseases.
Collapse
Affiliation(s)
- Kenly Wuputra
- Cell Therapy Research Center, Department of Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan; Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Regenerative Medicine and Cell Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Wen-Hung Hsu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Gangshan Hospital, Kaohsiung 820, Taiwan; Department of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Chia-Chen Ku
- Cell Therapy Research Center, Department of Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan; Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Regenerative Medicine and Cell Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Ya-Han Yang
- Division of General Surgery, E-DA Dachang Hospital, Kaohsiung 80706, Taiwan.
| | - Kung-Kai Kuo
- Division of General Surgery, E-DA Dachang Hospital, Kaohsiung 80706, Taiwan.
| | - Fang-Jung Yu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Gangshan Hospital, Kaohsiung 820, Taiwan; Department of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan.
| | - Hsin-Su Yu
- Emeritus Professor in College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| | - Kyosuke Nagata
- Professor, Insitutte of Medicine, University of Tsukuba, Tsukuba 3058577, Japan.
| | - Deng-Chyang Wu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Gangshan Hospital, Kaohsiung 820, Taiwan; Department of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Chao-Hung Kuo
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Gangshan Hospital, Kaohsiung 820, Taiwan; Department of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Superintendant in Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan.
| | - Kazunari K Yokoyama
- Cell Therapy Research Center, Department of Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan; Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Regenerative Medicine and Cell Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| |
Collapse
|
9
|
Del Sorbo L, Giugliano R, Cerracchio C, Iovane V, Salvatore MM, Serra F, Amoroso MG, Pellegrini F, Levante M, Capozza P, Diakoudi G, Galdiero M, Fusco G, Pratelli A, Andolfi A, Fiorito F. In Vitro Evaluation of Aryl Hydrocarbon Receptor Involvement in Feline Coronavirus Infection. Viruses 2025; 17:227. [PMID: 40006982 PMCID: PMC11860311 DOI: 10.3390/v17020227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/01/2025] [Accepted: 02/03/2025] [Indexed: 02/27/2025] Open
Abstract
Feline coronavirus (FCoV) is an alphacoronavirus (αCoV) that causes moderate or chronic asymptomatic infection in cats. However, in a single infected cat, FCoV can modify its cellular tropism by acquiring the ability to infect macrophages, resulting in the development of feline infectious peritonitis (FIP). In this context, to restrain the impact of FCoV infection, scientific research has focused attention on the development of antiviral therapies involving novel mechanisms of action. Recent studies have demonstrated that aryl hydrocarbon receptor (AhR) signaling regulates the host response to different human and animal CoVs. Hence, the mechanism of action of AhR was evaluated upon FCoV infection in Crandell Feline Kidney (CRFK) and in canine fibrosarcoma (A72) cells. Following infection with feline enteric CoV (FECV), strain "München", a significant activation of AhR and of its target CYP1A1, was observed. The selective AhR antagonist CH223191 provoked a reduction in FCoV replication and in the levels of viral nucleocapsid protein (NP). Furthermore, the effect of the AhR inhibitor on the acidity of lysosomes in infected cells was observed. Our findings indicate that FCoV acts on viral replication that upregulates AhR. CH223191 repressed virus yield through the inhibition of AhR. In this respect, for counteracting FCoV, AhR represents a new target useful for identifying antiviral drugs. Moreover, in the presence of CH223191, the alkalinization of lysosomes in FCoV-infected CRFK cells was detected, outlining their involvement in antiviral activity.
Collapse
Affiliation(s)
- Luca Del Sorbo
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy; (L.D.S.); (R.G.); (C.C.)
| | - Rosa Giugliano
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy; (L.D.S.); (R.G.); (C.C.)
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy;
| | - Claudia Cerracchio
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy; (L.D.S.); (R.G.); (C.C.)
| | - Valentina Iovane
- Department of Agricultural Sciences, University of Naples Federico II, 80055 Portici, Italy; (V.I.); (A.A.)
| | - Maria Michela Salvatore
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy; (L.D.S.); (R.G.); (C.C.)
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy
| | - Francesco Serra
- Istituto Zooprofilattico del Mezzogiorno, 80055 Portici, Italy; (F.S.); (M.G.A.); (M.L.)
| | - Maria Grazia Amoroso
- Istituto Zooprofilattico del Mezzogiorno, 80055 Portici, Italy; (F.S.); (M.G.A.); (M.L.)
| | - Francesco Pellegrini
- Department of Veterinary Medicine, University of Bari, 70010 Valenzan, Italy; (F.P.); (P.C.); (G.D.)
| | - Martina Levante
- Istituto Zooprofilattico del Mezzogiorno, 80055 Portici, Italy; (F.S.); (M.G.A.); (M.L.)
| | - Paolo Capozza
- Department of Veterinary Medicine, University of Bari, 70010 Valenzan, Italy; (F.P.); (P.C.); (G.D.)
| | - Georgia Diakoudi
- Department of Veterinary Medicine, University of Bari, 70010 Valenzan, Italy; (F.P.); (P.C.); (G.D.)
| | - Massimiliano Galdiero
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy;
| | - Giovanna Fusco
- Istituto Zooprofilattico del Mezzogiorno, 80055 Portici, Italy; (F.S.); (M.G.A.); (M.L.)
| | - Annamaria Pratelli
- Department of Veterinary Medicine, University of Bari, 70010 Valenzan, Italy; (F.P.); (P.C.); (G.D.)
| | - Anna Andolfi
- Department of Agricultural Sciences, University of Naples Federico II, 80055 Portici, Italy; (V.I.); (A.A.)
- BAT Center-Interuniversity Center for Studies on Bioinspired Agro-Environmental Technology, University of Naples Federico II, 80055 Portici, Italy
| | - Filomena Fiorito
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy; (L.D.S.); (R.G.); (C.C.)
- BAT Center-Interuniversity Center for Studies on Bioinspired Agro-Environmental Technology, University of Naples Federico II, 80055 Portici, Italy
| |
Collapse
|
10
|
Hsu J, Fang HH, Su JGJ. Telmisartan potentiates the ITE-induced aryl hydrocarbon receptor activity in human liver cell line. Arch Toxicol 2025; 99:667-678. [PMID: 39480548 DOI: 10.1007/s00204-024-03901-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 10/17/2024] [Indexed: 11/02/2024]
Abstract
Telmisartan is an angiotensin receptor blocker (ARB) approved by the Food and Drug Administration of the US for the treatment of hypertension. It possesses unique pharmacologic properties, including the longest half-life among all ARBs; this leads to a 24-h sustained reduction of blood pressure. Besides well-known antihypertensive and cardioprotective effects, there is also strong clinical evidence that telmisartan confers renoprotection. Aryl hydrocarbon receptor (AhR) belongs to the steroid receptor family. 2-(1'H-indole-3'-carbonyl)-thiazole-4-carboxylic acid methyl ester (ITE) is an endogenous ligand of AhR. Cytochrome P450 (CYP) 1A1 is an AhR-target gene. In this article, we demonstrated that telmisartan (2.5-60 μM) enhanced CYP1A1 promoter activity and expressions of mRNA and protein. Telmisartan-induced CYP1A1 expression was blocked by the AhR antagonist CH-223191 in liver cell lines and was negligible in the AhR signaling-deficient mutant cells. In addition, telmisartan induced transcriptional activity mediated by aryl hydrocarbon response element in both human and mouse cells, and was able to induce AhR translocation into the nucleus. Accordingly, telmisartan is an AhR agonist. It also acted synergistically with ITE to further enhance the expression of CYP1A1 mRNA and protein. This synergistic effect was more pronounced in cells with AhR overexpression compared to those without. AhR activity has strong association with the progression of chronic renal disease. Our study demonstrated that telmisartan is an AhR agonist and has synergistic effect with ITE, an indole derivative, to potentiate the effect on AhR. This finding may provide additional clues about the mechanism of the protective effect of telmisartan on the kidney.
Collapse
Affiliation(s)
- Jiun Hsu
- National Taiwan University Hospital Yunlin Branch, Yunlin, 640203, Taiwan, ROC
| | - Hsiao-Ho Fang
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi, 60004, Taiwan, ROC
| | - Jyan-Gwo Joseph Su
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi, 60004, Taiwan, ROC.
| |
Collapse
|
11
|
Chen NX, O’Neill KD, Wilson HE, Srinivasan S, Bonewald L, Moe SM. The uremic toxin indoxyl sulfate decreases osteocyte RANKL/OPG and increases Wnt inhibitor RNA expression that is reversed by PTH. JBMR Plus 2025; 9:ziae136. [PMID: 39664935 PMCID: PMC11631378 DOI: 10.1093/jbmrpl/ziae136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/04/2024] [Accepted: 10/27/2024] [Indexed: 12/13/2024] Open
Abstract
Renal osteodystrophy (ROD) leads to increased fractures, potentially due to underlying low bone turnover in chronic kidney disease (CKD). We hypothesized that indoxyl sulfate (IS), a circulating toxin elevated in CKD and a ligand for the aryl hydrocarbon receptor (AhR), may target the osteocytes leading to bone cell uncoupling in ROD. The IDG-SW3 osteocytes were cultured for 14 days (early) and 35 days (mature osteocytes) and incubated with 500 μM of IS after dose finding studies to confirm AhR activation. Long-term incubation of IS for 14 days led to decreased expression of Tnfsf11/Tnfrsf11b ratio (RANKL/OPG), which would increase osteoclast activity, and increased expression of Wnt inhibitors Sost and Dkk1, which would decrease bone formation in addition to decreased mineralization and alkaline phosphatase (ALP) activity. When osteocytes were incubated with IS and the AhR translocation inhibitor CH223191, mineralization and ALP activity were restored. However, the Tnfsf11/Tnfrsf11b ratio and Sost, Dkk1 expression were not altered compared with IS alone, suggesting more complex signaling. In both early and mature osteocytes, co-culture with parathyroid hormone (PTH) and IS reversed the IS-induced upregulation of Sost and Dkk1, and IS enhanced the PTH-induced increase of the Tnfsf11/Tnfrsf11b ratio. Co-culture of IS with PTH additively enhanced the AhR activity assessed by Cyp1a1 and Cyp1b1 expression. In summary, IS in the absence of PTH increased osteocyte messenger RNA (mRNA) Wnt inhibitor expression in both early and mature osteocytes, decreased mRNA expression ofTnfsf11/Tnfrsf11b ratio and decreased mineralization in early osteocytes. These changes would lead to decreased resorption and formation resulting in low bone remodeling. These data suggest IS may be important in the underlying low turnover bone disease observed in CKD when PTH is not elevated. In addition, when PTH is elevated, IS interacts to further increase Tnfsf11/Tnfrsf11b ratio for osteoclast activity in both early and mature osteocytes, which would worsen bone resorption.
Collapse
Affiliation(s)
- Neal X Chen
- Department of Medicine, Division of Nephrology and Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Kalisha D O’Neill
- Department of Medicine, Division of Nephrology and Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Hannah E Wilson
- Department of Anatomy, Cell Biology and Physiology Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Shruthi Srinivasan
- Department of Medicine, Division of Nephrology and Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Lynda Bonewald
- Department of Anatomy, Cell Biology and Physiology Indiana University School of Medicine, Indianapolis, IN 46202, United States
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Sharon M Moe
- Department of Medicine, Division of Nephrology and Indiana University School of Medicine, Indianapolis, IN 46202, United States
- Department of Anatomy, Cell Biology and Physiology Indiana University School of Medicine, Indianapolis, IN 46202, United States
| |
Collapse
|
12
|
Zhang L, Chen WQ, Han XY, Wang HL, Gao PZ, Wang DM, Cao Z, Sun CH, Cheng D, Bai J, He QL, Liu SZ. Benzo(a)pyrene exposure during pregnancy leads to germ cell apoptosis in male mice offspring via affecting histone modifications and oxidative stress levels. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 952:175877. [PMID: 39226951 DOI: 10.1016/j.scitotenv.2024.175877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 08/23/2024] [Accepted: 08/28/2024] [Indexed: 09/05/2024]
Abstract
Infertility has gradually become a global health concern, and evidence suggests that exposure to environmental endocrine-disrupting chemicals (EDCs) represent one of the key causes of infertility. Benzo(a)pyrene (BaP) is a typical EDC that is widespread in the environment. Previous studies have detected BaP in human urine, semen, cervical mucus, oocytes and follicular fluid, resulting in reduced fertility and irreversible reproductive damage. However, the mechanisms underlying the effects of gestational BaP exposure on offspring fertility in male mice have not been fully explored. In this study, pregnant mice were administered BaP at doses of 0, 5, 10 and 20 mg/kg/day via gavage from Days 7.5 to 12.5 of gestation. The results revealed that BaP exposure during pregnancy disrupted the structural integrity of testicular tissue, causing a disorganized arrangement of spermatogenic cells, compromised sperm quality, elevated levels of histone modifications and increased apoptosis in the testicular tissue of F1 male mice. Furthermore, oxidative stress was also increased in the testicular tissue of F1 male mice. BaP activated the AhR/ERα signaling pathway, affected H3K4me3 expression and induced apoptosis in testicular tissue. AhR and Cyp1a1 were overexpressed, and the expression of key molecules in the antioxidant pathway, including Keap1 and Nrf2, was reduced. The combined effects of these molecules led to apoptosis in testicular tissues, damaging and compromising sperm quality. This impairment in testicular cells further contributed to compromised testicular tissues, ultimately impacting the reproductive health of F1 male mice.
Collapse
Affiliation(s)
- Lin Zhang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China; Dongying Institute, Shandong Normal University, Dongying 257000, China
| | - Wen-Qi Chen
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China; Dongying Institute, Shandong Normal University, Dongying 257000, China
| | - Xiao-Ying Han
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China; Dongying Institute, Shandong Normal University, Dongying 257000, China
| | - Hong-Li Wang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China; Dongying Institute, Shandong Normal University, Dongying 257000, China
| | - Peng-Zhi Gao
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China; Dongying Institute, Shandong Normal University, Dongying 257000, China
| | - Dong-Mei Wang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China; Dongying Institute, Shandong Normal University, Dongying 257000, China
| | - Zheng Cao
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China; Dongying Institute, Shandong Normal University, Dongying 257000, China
| | - Chang-Hua Sun
- Division of Toxicology, Shandong Center for Disease Control and Prevention, Jinan 250014, China
| | - Dong Cheng
- Division of Toxicology, Shandong Center for Disease Control and Prevention, Jinan 250014, China
| | - Jing Bai
- Department of Maternity, Jinan Maternity and Child Care Hospital Affiliated to Shandong First Medical University, Jinan Maternity and Child Care Hospital, Jinan 250001, China.
| | - Qi-Long He
- Division of Toxicology, Shandong Center for Disease Control and Prevention, Jinan 250014, China.
| | - Shu-Zhen Liu
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China; Dongying Institute, Shandong Normal University, Dongying 257000, China.
| |
Collapse
|
13
|
Dexheimer TS, Coussens NP, Silvers T, Jones EM, Chen L, Fang J, Morris J, Moscow JA, Doroshow JH, Teicher BA. Combination screen in multi-cell type tumor spheroids reveals interaction between aryl hydrocarbon receptor antagonists and E1 ubiquitin-activating enzyme inhibitor. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2024; 29:100186. [PMID: 39362362 PMCID: PMC11562894 DOI: 10.1016/j.slasd.2024.100186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that regulates genes of drug transporters and metabolic enzymes to detoxify small molecule xenobiotics. It has a complex role in cancer biology, influencing both the progression and suppression of tumors by modulating malignant properties of tumor cells and anti-tumor immunity, depending on the specific tumor type and developmental stage. This has led to the discovery and development of selective AhR modulators, including BAY 2416964 which is currently in clinical trials. To identify small molecule anticancer agents that might be combined with AhR antagonists for cancer therapy, a high-throughput combination screen was performed using multi-cell type tumor spheroids grown from malignant cells, endothelial cells, and mesenchymal stem cells. The AhR selective antagonists BAY 2416964, GNF351, and CH-223191 were tested individually and in combination with twenty-five small molecule anticancer agents. As single agents, BAY 2416964 and CH-223191 showed minimal activity, whereas GNF351 reduced the viability of some spheroid models at concentrations greater than 1 µM. The activity of most combinations aligned well with the single agent activity of the combined agent, without apparent contributions from the AhR antagonist. All three AhR antagonists sensitized tumor spheroids to TAK-243, an E1 ubiquitin-activating enzyme inhibitor. These combinations were active in spheroids containing bladder, breast, ovary, kidney, pancreas, colon, and lung tumor cell lines. The AhR antagonists also potentiated pevonedistat, a selective inhibitor of the NEDD8-activating enzyme E1 regulatory subunit, in several tumor spheroid models. In contrast, the AhR antagonists did not enhance the cytotoxicity of the proteasome inhibitor bortezomib.
Collapse
Affiliation(s)
- Thomas S Dexheimer
- Target Validation and Screening Laboratory, Molecular Pharmacology Laboratory, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, 1050 Boyles Street, Frederick, MD 21702, USA.
| | - Nathan P Coussens
- Target Validation and Screening Laboratory, Molecular Pharmacology Laboratory, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, 1050 Boyles Street, Frederick, MD 21702, USA
| | - Thomas Silvers
- Target Validation and Screening Laboratory, Molecular Pharmacology Laboratory, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, 1050 Boyles Street, Frederick, MD 21702, USA
| | - Eric M Jones
- Target Validation and Screening Laboratory, Molecular Pharmacology Laboratory, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, 1050 Boyles Street, Frederick, MD 21702, USA
| | - Li Chen
- Molecular Characterization Laboratory, Clinical Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Jianwen Fang
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Joel Morris
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jeffrey A Moscow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - James H Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Beverly A Teicher
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
14
|
Xie H, Yang N, Lu L, Sun X, Li J, Wang X, Guo H, Zhou L, Liu J, Wu H, Yu C, Zhang W, Lu L. Uremic Toxin Receptor AhR Facilitates Renal Senescence and Fibrosis via Suppressing Mitochondrial Biogenesis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402066. [PMID: 38940381 PMCID: PMC11434102 DOI: 10.1002/advs.202402066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/02/2024] [Indexed: 06/29/2024]
Abstract
Retention of metabolic end-products in the bodily fluids of patients with chronic kidney disease (CKD) may lead to uremia. The uremic toxin indoxyl sulfate (IS), a tryptophan metabolite, is an endogenous ligand of aryl hydrocarbon receptor (AhR). It is clarified that the upregulation and activation of AhR by IS in tubular epithelial cells (TECs) promote renal senescence and fibrosis. Renal TEC-specific knockout of AhR attenuates renal senescence and fibrosis, as well as the suppression of PGC1α-mediated mitochondrial biogenesis in ischemia reperfusion (IR)- or IS-treated CKD mice kidneys. Overexpression of peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC1α) attenuates IS-induced cell senescence and extracellular matrix production in cultured TECs. Mechanistically, AhR is able to interact with PGC1α and promotes the ubiquitin degradation of PGC1α via its E3 ubiquitin ligase activity. In summary, the elevation and activation of AhR by the accumulated uremic toxins in the progression of CKD accelerate renal senescence and fibrosis by suppressing mitochondrial biogenesis via promoting ubiquitination and proteasomal degradation of PGC1α.
Collapse
Affiliation(s)
- Hongyan Xie
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
- Department of Nephrology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Ninghao Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Li Lu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Dali University, Dali, Yunnan, 671013, China
| | - Xi'ang Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Jingyao Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Xin Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Hengjiang Guo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Li Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Jun Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Huijuan Wu
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Chen Yu
- Department of Nephrology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Wei Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Limin Lu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Shanghai, 201102, China
| |
Collapse
|
15
|
Sukka SR, Ampomah PB, Darville LNF, Ngai D, Wang X, Kuriakose G, Xiao Y, Shi J, Koomen JM, McCusker RH, Tabas I. Efferocytosis drives a tryptophan metabolism pathway in macrophages to promote tissue resolution. Nat Metab 2024; 6:1736-1755. [PMID: 39242914 PMCID: PMC11734744 DOI: 10.1038/s42255-024-01115-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 07/24/2024] [Indexed: 09/09/2024]
Abstract
Macrophage efferocytosis prevents apoptotic cell (AC) accumulation and triggers inflammation-resolution pathways. The mechanisms linking efferocytosis to resolution often involve changes in macrophage metabolism, but many gaps remain in our understanding of these processes. We now report that efferocytosis triggers an indoleamine 2,3-dioxygenase-1 (IDO1)-dependent tryptophan (Trp) metabolism pathway that promotes several key resolution processes, including the induction of pro-resolving proteins, such interleukin-10, and further enhancement of efferocytosis. The process begins with upregulation of Trp transport and metabolism, and it involves subsequent activation of the aryl hydrocarbon receptor (AhR) by the Trp metabolite kynurenine (Kyn). Through these mechanisms, macrophage IDO1 and AhR contribute to a proper resolution response in several different mouse models of efferocytosis-dependent tissue repair, notably during atherosclerosis regression induced by plasma low-density lipoprotein (LDL) lowering. These findings reveal an integrated metabolism programme in macrophages that links efferocytosis to resolution, with possible therapeutic implications for non-resolving chronic inflammatory diseases, notably atherosclerosis.
Collapse
Affiliation(s)
- Santosh R Sukka
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| | - Patrick B Ampomah
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Lancia N F Darville
- Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - David Ngai
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Xiaobo Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - George Kuriakose
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Yuling Xiao
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - John M Koomen
- Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Robert H McCusker
- Department of Animal Sciences, Integrative Immunology and Behavior Program and Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Ira Tabas
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
- Departments of Physiology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
16
|
Wang D, Zhang J, Yin H, Yan R, Wang Z, Deng J, Li G, Pan Y. The anti-tumor effects of cosmosiin through regulating AhR/CYP1A1-PPARγ in breast cancer. FASEB J 2024; 38:e70002. [PMID: 39162680 DOI: 10.1096/fj.202401191r] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/21/2024] [Accepted: 08/07/2024] [Indexed: 08/21/2024]
Abstract
Breast cancer is one of the threatening malignant tumors with the highest mortality and incidence rate over the world. There are a lot of breast cancer patients dying every year due to the lack of effective and safe therapeutic drugs. Therefore, it is highly necessary to develop more effective drugs to overcome breast cancer. As a glycoside derivative of apigenin, cosmosiin is characterized by low toxicity, high water solubility, and wide distribution in nature. Additionally, cosmosiin has been shown to perform anti-tumor effects in cervical cancer, hepatocellular carcinoma and melanoma. However, its pharmacological effects on breast cancer and its mechanisms are still unknown. In our study, the anti-breast cancer effect and mechanism of cosmosiin were investigated by using breast cancer models in vivo and in vitro. The results showed that cosmosiin inhibited the proliferation, migration, and adhesion of breast cancer cells in vitro and suppressed the growth of tumor in vivo through binding with AhR and inhibiting it, thus regulating the downstream CYP1A1/AMPK/mTOR and PPARγ/Wnt/β-catenin signaling pathways. Collectively, our findings have made contribution to the development of novel drugs against breast cancer by targeting AhR and provided a new direction for the research in the field of anti-breast cancer therapy.
Collapse
Affiliation(s)
- Dan Wang
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
| | - Jing Zhang
- Inner Mongolia Key Laboratory of Molecular Biology, Inner Mongolia Medical University, Hohhot, China
| | - Houqing Yin
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
| | - Ribai Yan
- School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Zequn Wang
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
| | - Jinhai Deng
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | - Gang Li
- Department of General Surgery, Peking University Third Hospital, Beijing, China
| | - Yan Pan
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
- Beijing Key Laboratory of Tumor Systems Biology, Peking University, Beijing, China
| |
Collapse
|
17
|
Yasuda T, Takagi T, Asaeda K, Hashimoto H, Kajiwara M, Azuma Y, Kitae H, Hirai Y, Mizushima K, Doi T, Inoue K, Dohi O, Yoshida N, Uchiyama K, Ishikawa T, Konishi H, Ukawa Y, Kohara A, Kudoh M, Inoue R, Naito Y, Itoh Y. Urolithin A-mediated augmentation of intestinal barrier function through elevated secretory mucin synthesis. Sci Rep 2024; 14:15706. [PMID: 38977770 PMCID: PMC11231190 DOI: 10.1038/s41598-024-65791-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 06/24/2024] [Indexed: 07/10/2024] Open
Abstract
Maintaining the mucus layer is crucial for the innate immune system. Urolithin A (Uro A) is a gut microbiota-derived metabolite; however, its effect on mucin production as a physical barrier remains unclear. This study aimed to elucidate the protective effects of Uro A on mucin production in the colon. In vivo experiments employing wild-type mice, NF-E2-related factor 2 (Nrf2)-deficient mice, and wild-type mice treated with an aryl hydrocarbon receptor (AhR) antagonist were conducted to investigate the physiological role of Uro A. Additionally, in vitro assays using mucin-producing cells (LS174T) were conducted to assess mucus production following Uro A treatment. We found that Uro A thickened murine colonic mucus via enhanced mucin 2 expression facilitated by Nrf2 and AhR signaling without altering tight junctions. Uro A reduced mucosal permeability in fluorescein isothiocyanate-dextran experiments and alleviated dextran sulfate sodium-induced colitis. Uro A treatment increased short-chain fatty acid-producing bacteria and propionic acid concentration. LS174T cell studies confirmed that Uro A promotes mucus production through the AhR and Nrf2 pathways. In conclusion, the enhanced intestinal mucus secretion induced by Uro A is mediated through the actions of Nrf-2 and AhR, which help maintain intestinal barrier function.
Collapse
Affiliation(s)
- Takeshi Yasuda
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Tomohisa Takagi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan.
- Department for Medical Innovation and Translational Medical Science, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan.
| | - Kohei Asaeda
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Hikaru Hashimoto
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Mariko Kajiwara
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Yuka Azuma
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Hiroaki Kitae
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Yasuko Hirai
- Department of Human Immunology and Nutrition Science, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Katsura Mizushima
- Department of Human Immunology and Nutrition Science, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Toshifumi Doi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Ken Inoue
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Osamu Dohi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Naohisa Yoshida
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Kazuhiko Uchiyama
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Takeshi Ishikawa
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Hideyuki Konishi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Yuichi Ukawa
- Daicel Corporation, Healthcare SBU, Tokyo, 108-8230, Japan
| | - Akiko Kohara
- Daicel Corporation, Healthcare SBU, Tokyo, 108-8230, Japan
| | - Masatake Kudoh
- Daicel Corporation, Healthcare SBU, Niigata, 944-8550, Japan
| | - Ryo Inoue
- Laboratory of Animal Science, Department of Applied Biological Sciences, Faculty of Agriculture, Setsunan University, Hirakata, 572-8508, Japan
| | - Yuji Naito
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
- Department of Human Immunology and Nutrition Science, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Yoshito Itoh
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| |
Collapse
|
18
|
Boychenko S, Egorova VS, Brovin A, Egorov AD. White-to-Beige and Back: Adipocyte Conversion and Transcriptional Reprogramming. Pharmaceuticals (Basel) 2024; 17:790. [PMID: 38931457 PMCID: PMC11206576 DOI: 10.3390/ph17060790] [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: 05/21/2024] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
Obesity has become a pandemic, as currently more than half a billion people worldwide are obese. The etiology of obesity is multifactorial, and combines a contribution of hereditary and behavioral factors, such as nutritional inadequacy, along with the influences of environment and reduced physical activity. Two types of adipose tissue widely known are white and brown. While white adipose tissue functions predominantly as a key energy storage, brown adipose tissue has a greater mass of mitochondria and expresses the uncoupling protein 1 (UCP1) gene, which allows thermogenesis and rapid catabolism. Even though white and brown adipocytes are of different origin, activation of the brown adipocyte differentiation program in white adipose tissue cells forces them to transdifferentiate into "beige" adipocytes, characterized by thermogenesis and intensive lipolysis. Nowadays, researchers in the field of small molecule medicinal chemistry and gene therapy are making efforts to develop new drugs that effectively overcome insulin resistance and counteract obesity. Here, we discuss various aspects of white-to-beige conversion, adipose tissue catabolic re-activation, and non-shivering thermogenesis.
Collapse
Affiliation(s)
- Stanislav Boychenko
- Gene Therapy Department, Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sirius, Russia; (S.B.); (A.B.)
| | - Vera S. Egorova
- Biotechnology Department, Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sirius, Russia
| | - Andrew Brovin
- Gene Therapy Department, Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sirius, Russia; (S.B.); (A.B.)
| | - Alexander D. Egorov
- Gene Therapy Department, Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sirius, Russia; (S.B.); (A.B.)
| |
Collapse
|
19
|
Kumari S, Singh B, Kureel AK, Saini S, Prakash S, Chauhan A, Kumar P, Singh K, Rai AK. Benzo[a]pyrene exposure causes exonal switch resulting in reduced surface CD5 expression in an AHR-dependent manner. Immunol Lett 2024; 267:106858. [PMID: 38631465 DOI: 10.1016/j.imlet.2024.106858] [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: 06/17/2023] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 04/19/2024]
Abstract
The function of CD5 protein in T cells is well documented, but regulation of its surface-level expression has yet to be fully understood. However, variation in its surface expression is associated with various immunopathological conditions and haematological malignancies. Briefly, expression of an alternate exon E1B of a human endogenous retroviruses (HERV) origin directly downregulates the conventional transcript variant (E1A), as its expression leads to the retention of the resultant protein at the intracellular level (cCD5). A separate promoter governs the expression of E1B and may be influenced by different transcription factors. Hence, we performed in silico transcription factor binding site (TFBS) analysis of the 3 kb upstream region from TSS of exon E1B and found five putative DREs (Dioxin Response elements) with good similarity scores. Further, we observed the upregulation in E1B expression after the exposure of BaP (a dioxin) and the reduction of E1A expression and their respective protein, i.e. sCD5 and cCD5. The binding of AHR at the predicted DRE sites was confirmed by ChIP qPCR and AHR specific inhibitor and gene silencing studies suggested the involvement of AHR in exonal switch. This study indicates that the polycyclic aromatic hydrocarbon decreases the sCD5 expression by upregulating alternative exon expression, which may adversely affect the overall T cell functions.
Collapse
Affiliation(s)
- Smita Kumari
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, U.P. 211004, India
| | - Bharat Singh
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, U.P. 211004, India
| | - Amit Kumar Kureel
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, U.P. 211004, India
| | - Sheetal Saini
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, U.P. 211004, India
| | - Satya Prakash
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, U.P. 211004, India
| | - Aditi Chauhan
- Stem Cell Research Center, Department of Hematology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, U.P. 226014, India
| | - Prabin Kumar
- Department of Transplant Immunology and Immunogenetics, All India Institute of medical Sciences (A.I.I.M.S.), New Delhi, 110029, India
| | - Kulwant Singh
- Stem Cell Research Center, Department of Hematology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, U.P. 226014, India
| | - Ambak Kumar Rai
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, U.P. 211004, India.
| |
Collapse
|
20
|
Ma S, Wu Q, Wu W, Tian Y, Zhang J, Chen C, Sheng X, Zhao F, Ding L, Wang T, Zhao L, Xie Y, Wang Y, Yue X, Wu Z, Wei J, Zhang K, Liang X, Gao L, Wang H, Wang G, Li C, Ma C. Urolithin A Hijacks ERK1/2-ULK1 Cascade to Improve CD8 + T Cell Fitness for Antitumor Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2310065. [PMID: 38447147 PMCID: PMC11095213 DOI: 10.1002/advs.202310065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/20/2024] [Indexed: 03/08/2024]
Abstract
According to the latest evidence, the microbial metabolite Urolithin A (UA), known for its role in promoting cellular health, modulates CD8+ T cell-mediated antitumor activity. However, the direct target protein of UA and its underlying mechanism remains unclear. Here, this research identifies ERK1/2 as the specific target crucial for UA-mediated CD8+ T cell activation. Even at low doses, UA markedly enhances the persistence and effector functions of primary CD8+ cytotoxic T lymphocytes (CTLs) and human chimeric antigen receptor (CAR) T cells both in vitro and in vivo. Mechanistically, UA interacts directly with ERK1/2 kinases, enhancing their activation and subsequently facilitating T cell activation by engaging ULK1. The UA-ERK1/2-ULK1 axis promotes autophagic flux in CD8+ CTLs, enhancing cellular metabolism and maintaining reactive oxygen species (ROS) levels, as evidenced by increased oxygen consumption and extracellular acidification rates. UA-treated CD8+ CTLs also display elevated ATP levels and enhanced spare respiratory capacity. Overall, UA activates ERK1/2, inducing autophagy and metabolic adaptation, showcasing its potential in tumor immunotherapy and interventions for diseases involving ERKs.
Collapse
Affiliation(s)
- Shuaiya Ma
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Sciences, Qilu Hospital, Cheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
| | - Qi Wu
- GI Cancer Research InstituteTongji HospitalHuazhong University of Science and TechnologyWuhanHubei430074P. R. China
| | - Wenxian Wu
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Sciences, Qilu Hospital, Cheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
- Guangdong Key Laboratory of Age‐Related Cardiac and Cerebral DiseaseDepartment of NeurologyAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong524001P. R. China
- Shenzhen Research Institute of Shandong UniversityShenzhen518057P. R. China
| | - Ye Tian
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Sciences, Qilu Hospital, Cheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
| | - Jie Zhang
- Advanced Medical Research InstituteCheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
| | - Chaojia Chen
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Sciences, Qilu Hospital, Cheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
| | - Xue Sheng
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Sciences, Qilu Hospital, Cheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
| | - Fangcheng Zhao
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Sciences, Qilu Hospital, Cheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
| | - Lu Ding
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Sciences, Qilu Hospital, Cheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
| | - Taixia Wang
- Central LaboratoryTongji University School of MedicineTongji UniversityShanghai200072P. R. China
| | - Laixi Zhao
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Sciences, Qilu Hospital, Cheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
| | - Yuying Xie
- Guangdong Key Laboratory of Age‐Related Cardiac and Cerebral DiseaseDepartment of NeurologyAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong524001P. R. China
| | - Yongxiang Wang
- Guangdong Key Laboratory of Age‐Related Cardiac and Cerebral DiseaseDepartment of NeurologyAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong524001P. R. China
| | - Xuetian Yue
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Cell BiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
| | - Zhuanchang Wu
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Sciences, Qilu Hospital, Cheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
| | - Jian Wei
- Department of PhysiologySchool of Basic Medical SciencesShandong UniversityJinan250012P. R. China
| | - Kun Zhang
- Central LaboratoryTongji University School of MedicineTongji UniversityShanghai200072P. R. China
| | - Xiaohong Liang
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Sciences, Qilu Hospital, Cheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
| | - Lifen Gao
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Sciences, Qilu Hospital, Cheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
| | - Hongyan Wang
- State Key Laboratory of Cell BiologyShanghai Institute of Biochemistry and Cell BiologyCenter for Excellence in Molecular Cell ScienceChinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghai200031P. R. China
| | - Guihua Wang
- GI Cancer Research InstituteTongji HospitalHuazhong University of Science and TechnologyWuhanHubei430074P. R. China
| | - Chunyang Li
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Histology and EmbryologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
| | - Chunhong Ma
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Sciences, Qilu Hospital, Cheeloo College of MedicineShandong UniversityJinanShandong250012P. R. China
| |
Collapse
|
21
|
Dos Santos IL, Mitchell M, Nogueira PAS, Lafita-Navarro MC, Perez-Castro L, Eriom J, Kilgore JA, Williams NS, Guo L, Xu L, Conacci-Sorrell M. Targeting of neuroblastoma cells through Kynurenine-AHR pathway inhibition. FEBS J 2024; 291:2172-2190. [PMID: 38431776 DOI: 10.1111/febs.17109] [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: 10/19/2023] [Revised: 01/13/2024] [Accepted: 02/21/2024] [Indexed: 03/05/2024]
Abstract
Neuroblastoma poses significant challenges in clinical management. Despite its relatively low incidence, this malignancy contributes disproportionately to cancer-related childhood mortality. Tailoring treatments based on risk stratification, including MYCN oncogene amplification, remains crucial, yet high-risk cases often confront therapeutic resistance and relapse. Here, we explore the aryl hydrocarbon receptor (AHR), a versatile transcription factor implicated in diverse physiological functions such as xenobiotic response, immune modulation, and cell growth. Despite its varying roles in malignancies, AHR's involvement in neuroblastoma remains elusive. Our study investigates the interplay between AHR and its ligand kynurenine (Kyn) in neuroblastoma cells. Kyn is generated from tryptophan (Trp) by the activity of the enzymes indoleamine 2,3-dioxygenase 1 (IDO1) and tryptophan 2,3-dioxygenase (TDO2). We found that neuroblastoma cells displayed sensitivity to the TDO2 inhibitor 680C91, exposing potential vulnerabilities. Furthermore, combining TDO2 inhibition with retinoic acid or irinotecan (two chemotherapeutic agents used to treat neuroblastoma patients) revealed synergistic effects in select cell lines. Importantly, clinical correlation analysis using patient data established a link between elevated expression of Kyn-AHR pathway genes and adverse prognosis, particularly in older children. These findings underscore the significance of the Kyn-AHR pathway in neuroblastoma progression, emphasizing its potential role as a therapeutic target.
Collapse
MESH Headings
- Humans
- Kynurenine/metabolism
- Neuroblastoma/pathology
- Neuroblastoma/metabolism
- Neuroblastoma/genetics
- Neuroblastoma/drug therapy
- Receptors, Aryl Hydrocarbon/metabolism
- Receptors, Aryl Hydrocarbon/genetics
- Receptors, Aryl Hydrocarbon/antagonists & inhibitors
- Cell Line, Tumor
- Tryptophan Oxygenase/metabolism
- Tryptophan Oxygenase/genetics
- Tryptophan Oxygenase/antagonists & inhibitors
- Tretinoin/pharmacology
- Signal Transduction/drug effects
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors
- Cell Proliferation/drug effects
- Gene Expression Regulation, Neoplastic/drug effects
Collapse
Affiliation(s)
- Igor Lopes Dos Santos
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Michael Mitchell
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Pediatrics, University of Texas Dell Medical School, Austin, TX, USA
| | - Pedro A S Nogueira
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - M Carmen Lafita-Navarro
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lizbeth Perez-Castro
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joyane Eriom
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jessica A Kilgore
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Noelle S Williams
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lei Guo
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lin Xu
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Maralice Conacci-Sorrell
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
22
|
Liu Y, Jin X, Ye Y, Xu Z, Du Z, Hong H, Yu H, Lin H, Huang X, Sun H. Emerging disinfection byproducts 3-bromine carbazole induces cardiac developmental toxicity via aryl hydrocarbon receptor activation in zebrafish larvae. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 346:123609. [PMID: 38395134 DOI: 10.1016/j.envpol.2024.123609] [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: 01/11/2024] [Revised: 02/05/2024] [Accepted: 02/17/2024] [Indexed: 02/25/2024]
Abstract
3-bromine carbazole (3-BCZ) represents a group of emerging aromatic disinfection byproducts (DBP) detected in drinking water; however, limited information is available regarding its potential cardiotoxicity. To assess its impacts, zebrafish embryos were exposed to 0, 0.06, 0.14, 0.29, 0.58, 1.44 or 2.88 mg/L of 3-BCZ for 120 h post fertilization (hpf). Our results revealed that ≥1.44 mg/L 3-BCZ exposure induced a higher incidence of heart malformation and an elevated pericardial area in zebrafish larvae; it also decreased the number of cardiac muscle cells and thins the walls of the ventricle and atrium while increasing cardiac output and impeding cardiac looping. Furthermore, 3-BCZ exposure also exhibited significant effects on the transcriptional levels of genes related to both cardiac development (nkx2.5, vmhc, gata4, tbx5, tbx2b, bmp4, bmp10, and bmp2b) and cardiac function (cacna1ab, cacna1da, atp2a1l, atp1b2b, atp1a3b, and tnnc1a). Notably, N-acetyl-L-cysteine, a reactive oxygen species scavenger, may alleviate the failure of cardiac looping induced by 3-BCZ but not the associated cardiac dysfunction or malformation; conversely, the aryl hydrocarbon receptor agonist CH131229 can completely eliminate the cardiotoxicity caused by 3-BCZ. This study provides new evidence for potential risks associated with ingesting 3-BCZ as well as revealing underlying mechanisms responsible for its cardiotoxic effects on zebrafish embryos.
Collapse
Affiliation(s)
- Yingying Liu
- Key Laboratory of Watershed Earth Surface Processes and Ecological Security, College of Geography and Environmental Science, Zhejiang Normal University, Jinhua, Zhejiang, 321004, PR China
| | - Xudong Jin
- Key Laboratory of Watershed Earth Surface Processes and Ecological Security, College of Geography and Environmental Science, Zhejiang Normal University, Jinhua, Zhejiang, 321004, PR China
| | - Yanan Ye
- Key Laboratory of Watershed Earth Surface Processes and Ecological Security, College of Geography and Environmental Science, Zhejiang Normal University, Jinhua, Zhejiang, 321004, PR China
| | - Zeqiong Xu
- Key Laboratory of Watershed Earth Surface Processes and Ecological Security, College of Geography and Environmental Science, Zhejiang Normal University, Jinhua, Zhejiang, 321004, PR China
| | - Zhongkun Du
- College of Resources and Environment, Shandong Agricultural University, Key Laboratory of Agricultural Environment in Universities of Shandong, 61 Daizong Road, Taian, 271018, PR China
| | - Huachang Hong
- Key Laboratory of Watershed Earth Surface Processes and Ecological Security, College of Geography and Environmental Science, Zhejiang Normal University, Jinhua, Zhejiang, 321004, PR China
| | - Haiying Yu
- Key Laboratory of Watershed Earth Surface Processes and Ecological Security, College of Geography and Environmental Science, Zhejiang Normal University, Jinhua, Zhejiang, 321004, PR China
| | - Hongjun Lin
- Key Laboratory of Watershed Earth Surface Processes and Ecological Security, College of Geography and Environmental Science, Zhejiang Normal University, Jinhua, Zhejiang, 321004, PR China
| | - Xianfeng Huang
- National and Local Joint Engineering Research Center for Ecological Treatment Technology of Urban Water Pollution, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, PR China
| | - Hongjie Sun
- Key Laboratory of Watershed Earth Surface Processes and Ecological Security, College of Geography and Environmental Science, Zhejiang Normal University, Jinhua, Zhejiang, 321004, PR China.
| |
Collapse
|
23
|
Tang JS, Stephens R, Li Y, Cait A, Gell K, Faulkner S, Grooby A, Herst PM, O'Sullivan D, Gasser O. Polyphenol and glucosinolate-derived AhR modulators regulate GPR15 expression on human CD4+ T cells. J Nutr Biochem 2023; 122:109456. [PMID: 37788725 DOI: 10.1016/j.jnutbio.2023.109456] [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: 07/27/2022] [Revised: 08/24/2023] [Accepted: 09/27/2023] [Indexed: 10/05/2023]
Abstract
Diets high in fruit and vegetables are perceived to be beneficial for intestinal homeostasis, in health as well as in the context of inflammatory bowel diseases (IBDs). Recent breakthroughs in the field of immunology have highlighted the importance of the ligand-activated transcription factor aryl hydrocarbon receptor (AhR) as a critical regulator of mucosal immunity, including the intestinal trafficking of CD4+ helper T cells, an immune cell subset implicated in a wide range of homeostatic and pathogenic processes. Specifically, the AhR has been shown to directly regulate the expression of the chemoattractant receptor G Protein-Coupled Receptor 15 (GPR15) on CD4+ T cells. GPR15 is an important gut homing marker whose expression on CD4+ T cells in the peripheral circulation is elevated in patients suffering from ulcerative colitis, raising the possibility that, in this setting, the beneficial effect of a diet rich in fruits and vegetables may be mediated through the modulation of GPR15 expression. To address this, we screened physiologically-relevant polyphenol and glucosinolate metabolites for their ability to affect both AhR activity and GPR15 expression. Our complementary approach and associated findings suggest that polyphenol and glucosinolate metabolites can regulate GPR15 expression on human CD4+ T cells in an AhR-dependent manner.
Collapse
Affiliation(s)
- Jeffry S Tang
- Malaghan Institute of Medical Research, Wellington, New Zealand; High-Value Nutrition National Science Challenge, Auckland, New Zealand.
| | - Ruth Stephens
- Malaghan Institute of Medical Research, Wellington, New Zealand; High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Yanyan Li
- Malaghan Institute of Medical Research, Wellington, New Zealand; High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Alissa Cait
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Katie Gell
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Sophie Faulkner
- Malaghan Institute of Medical Research, Wellington, New Zealand; High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Alix Grooby
- Malaghan Institute of Medical Research, Wellington, New Zealand; High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Patries M Herst
- Malaghan Institute of Medical Research, Wellington, New Zealand; Department of Radiation Therapy, University of Otago, Wellington, New Zealand
| | - David O'Sullivan
- Malaghan Institute of Medical Research, Wellington, New Zealand; High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Olivier Gasser
- Malaghan Institute of Medical Research, Wellington, New Zealand; High-Value Nutrition National Science Challenge, Auckland, New Zealand.
| |
Collapse
|
24
|
Chaudhry KA, Jacobi JJ, Gillard BM, Karasik E, Martin JC, da Silva Fernandes T, Hurley E, Feltri ML, Attwood KM, Twist CJ, Smiraglia DJ, Long MD, Bianchi-Smiraglia A. Aryl hydrocarbon receptor is a tumor promoter in MYCN-amplified neuroblastoma cells through suppression of differentiation. iScience 2023; 26:108303. [PMID: 38026169 PMCID: PMC10654598 DOI: 10.1016/j.isci.2023.108303] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/25/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Neuroblastoma is the most common extracranial solid tumor in children. MYCN amplification is detected in almost half of high-risk cases and is associated with poorly differentiated tumors, poor patient prognosis and poor response to therapy, including retinoids. We identify the aryl hydrocarbon receptor (AhR) as a transcription factor promoting the growth and suppressing the differentiation of MYCN-amplified neuroblastoma. A neuroblastoma specific AhR transcriptional signature reveals an inverse correlation of AhR activity with patients' outcome, suggesting AhR activity is critical for disease progression. AhR modulates chromatin structures, reducing accessibility to regions responsive to retinoic acid. Genetic and pharmacological inhibition of AhR results in induction of differentiation. Importantly, AhR antagonism with clofazimine synergizes with retinoic acid in inducing differentiation both in vitro and in vivo. Thus, we propose AhR as a target for MYCN-amplified neuroblastoma and that its antagonism, combined with current standard-of-care, may result in a more durable response in patients.
Collapse
Affiliation(s)
- Kanita A. Chaudhry
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Justine J. Jacobi
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Bryan M. Gillard
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Ellen Karasik
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Jeffrey C. Martin
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | | | - Edward Hurley
- Department of Biochemistry and Neurology, Institute for Myelin and Glia Exploration, State University of New York at Buffalo, Buffalo, NY, USA
| | - Maria Laura Feltri
- Department of Biochemistry and Neurology, Institute for Myelin and Glia Exploration, State University of New York at Buffalo, Buffalo, NY, USA
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Foundation I.R.C.C.S. Carlo Besta Neurological Institute Milan, Italy
| | - Kristopher M. Attwood
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Clare J. Twist
- Department of Pediatric Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Dominic J. Smiraglia
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Mark D. Long
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Anna Bianchi-Smiraglia
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
25
|
Wojciech L, Png CW, Koh EY, Kioh DYQ, Deng L, Wang Z, Wu L, Hamidinia M, Tung DWH, Zhang W, Pettersson S, Chan ECY, Zhang Y, Tan KSW, Gascoigne NRJ. A tryptophan metabolite made by a gut microbiome eukaryote induces pro-inflammatory T cells. EMBO J 2023; 42:e112963. [PMID: 37743772 PMCID: PMC10620759 DOI: 10.15252/embj.2022112963] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 08/11/2023] [Accepted: 09/06/2023] [Indexed: 09/26/2023] Open
Abstract
The large intestine harbors microorganisms playing unique roles in host physiology. The beneficial or detrimental outcome of host-microbiome coexistence depends largely on the balance between regulators and responder intestinal CD4+ T cells. We found that ulcerative colitis-like changes in the large intestine after infection with the protist Blastocystis ST7 in a mouse model are associated with reduction of anti-inflammatory Treg cells and simultaneous expansion of pro-inflammatory Th17 responders. These alterations in CD4+ T cells depended on the tryptophan metabolite indole-3-acetaldehyde (I3AA) produced by this single-cell eukaryote. I3AA reduced the Treg subset in vivo and iTreg development in vitro by modifying their sensing of TGFβ, concomitantly affecting recognition of self-flora antigens by conventional CD4+ T cells. Parasite-derived I3AA also induces over-exuberant TCR signaling, manifested by increased CD69 expression and downregulation of co-inhibitor PD-1. We have thus identified a new mechanism dictating CD4+ fate decisions. The findings thus shine a new light on the ability of the protist microbiome and tryptophan metabolites, derived from them or other sources, to modulate the adaptive immune compartment, particularly in the context of gut inflammatory disorders.
Collapse
Affiliation(s)
- Lukasz Wojciech
- Immunology Translational Research Programme, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Chin Wen Png
- Immunology Translational Research Programme, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Immunology Programme, Life Sciences InstituteNational University of SingaporeSingaporeSingapore
| | - Eileen Y Koh
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Dorinda Yan Qin Kioh
- Department of Pharmacy, Faculty of ScienceNational University of SingaporeSingaporeSingapore
| | - Lei Deng
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Ziteng Wang
- Department of Pharmacy, Faculty of ScienceNational University of SingaporeSingaporeSingapore
| | - Liang‐zhe Wu
- Immunology Translational Research Programme, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Maryam Hamidinia
- Immunology Translational Research Programme, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Desmond WH Tung
- Immunology Translational Research Programme, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Wei Zhang
- ASEAN Microbiome Nutrition CentreNational Neuroscience InstituteSingaporeSingapore
| | - Sven Pettersson
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- ASEAN Microbiome Nutrition CentreNational Neuroscience InstituteSingaporeSingapore
- Faculty of Medical SciencesSunway UniversitySubang JayaMalaysia
- Department of OdontologyKarolinska InstitutetStockholmSweden
| | - Eric Chun Yong Chan
- Department of Pharmacy, Faculty of ScienceNational University of SingaporeSingaporeSingapore
| | - Yongliang Zhang
- Immunology Translational Research Programme, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Immunology Programme, Life Sciences InstituteNational University of SingaporeSingaporeSingapore
| | - Kevin SW Tan
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Nicholas RJ Gascoigne
- Immunology Translational Research Programme, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- ASEAN Microbiome Nutrition CentreNational Neuroscience InstituteSingaporeSingapore
| |
Collapse
|
26
|
Khan DMIO, Karmaus PWF, Bach A, Crawford RB, Kaminski NE. An in vitro model of human hematopoiesis identifies a regulatory role for the aryl hydrocarbon receptor. Blood Adv 2023; 7:6253-6265. [PMID: 37477592 PMCID: PMC10589788 DOI: 10.1182/bloodadvances.2023010169] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/22/2023] [Accepted: 07/12/2023] [Indexed: 07/22/2023] Open
Abstract
In vitro models to study simultaneous development of different human immune cells and hematopoietic lineages are lacking. We identified and characterized, using single-cell methods, an in vitro stromal cell-free culture system of human hematopoietic stem and progenitor cell (HSPC) differentiation that allows concurrent development of multiple immune cell lineages. The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor influencing many biological processes in diverse cell types. Using this in vitro model, we found that AHR activation by the highly specific AHR ligand, 2,3,7,8-tetrachlorodibenzo-p-dioxin, drives differentiation of human umbilical cord blood-derived CD34+ HSPCs toward monocytes and granulocytes with a significant decrease in lymphoid and megakaryocyte lineage specification that may lead to reduced immune competence. To our knowledge, we also discovered for the first time, using single-cell modalities, that AHR activation decreased the expression of BCL11A and IRF8 in progenitor cells, which are critical genes involved in hematopoietic lineage specification processes at both transcriptomic and protein levels. Our in vitro model of hematopoiesis, coupled with single-cell tools, therefore allows for a better understanding of the role played by AHR in modulating hematopoietic differentiation.
Collapse
Affiliation(s)
- D M Isha Olive Khan
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI
| | - Peer W. F. Karmaus
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI
- National Institute of Environmental Health Sciences, Durham, NC
| | - Anthony Bach
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI
| | - Robert B. Crawford
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI
| | - Norbert E. Kaminski
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI
| |
Collapse
|
27
|
Li K, Li K, He Y, Liang S, Shui X, Lei W. Aryl hydrocarbon receptor: A bridge linking immuno-inflammation and metabolism in atherosclerosis. Biochem Pharmacol 2023; 216:115744. [PMID: 37579858 DOI: 10.1016/j.bcp.2023.115744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 08/16/2023]
Abstract
Cardiovascular disease is the leading cause of death worldwide, and atherosclerosis is a major contributor to this etiology. The ligand-activated transcription factor, known as the aryl hydrocarbon receptor (AhR), plays an essential role in the interactions between genes and the environment. In a number of human diseases, including atherosclerosis, the AhR signaling pathway has recently been shown to be aberrantly expressed and activated. It's reported that AhR can regulate the immuno-inflammatory response and metabolism pathways in atherosclerosis, potentially serving as a bridge that links these processes. In this review, we highlight the involvement of AhR in atherosclerosis. From the literature, we conclude that AhR is a potential target for controlling atherosclerosis through precise interventions.
Collapse
Affiliation(s)
- Kongwei Li
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Department of Precision Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Kaiyue Li
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Department of Precision Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yuan He
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Shan Liang
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Department of Precision Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Xiaorong Shui
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Laboratory of Vascular Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China.
| | - Wei Lei
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Department of Precision Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China.
| |
Collapse
|
28
|
Alluli A, Rijnbout St James W, Eidelman DH, Baglole CJ. Dynamic relationship between the aryl hydrocarbon receptor and long noncoding RNA balances cellular and toxicological responses. Biochem Pharmacol 2023; 216:115745. [PMID: 37597813 DOI: 10.1016/j.bcp.2023.115745] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/10/2023] [Accepted: 08/11/2023] [Indexed: 08/21/2023]
Abstract
The aryl hydrocarbon receptor (AhR) is a cytosolic transcription factor activated by endogenous ligands and xenobiotic chemicals. Once the AhR is activated, it translocates to the nucleus, dimerizes with the AhR nuclear translator (ARNT) and binds to xenobiotic response elements (XRE) to promote gene transcription, notably the cytochrome P450 CYP1A1. The AhR not only mediates the toxic effects of environmental chemicals, but also has numerous putative physiological functions. This dichotomy in AhR biology may be related to reciprocal regulation of long non-coding RNA (lncRNA). lncRNA are defined as transcripts more than 200 nucleotides in length that do not encode a protein but are implicated in many physiological processes such as cell differentiation, cell proliferation, and apoptosis. lncRNA are also linked to disease pathogenesis, particularly the development of cancer. Recent studies have revealed that AhR activation by environmental chemicals affects the expression and function of lncRNA. In this article, we provide an overview of AhR signaling pathways activated by diverse ligands and highlight key differences in the putative biological versus toxicological response of AhR activation. We also detail the functions of lncRNA and provide current data on their regulation by the AhR. Finally, we outline how overlap in function between AhR and lncRNA may be one way in which AhR can be both a regulator of endogenous functions but also a mediator of toxicological responses to environmental chemicals. Overall, more research is still needed to fully understand the dynamic interplay between the AhR and lncRNA.
Collapse
Affiliation(s)
- Aeshah Alluli
- Meakins-Christie Laboratories, McGill University, Canada; Translational Research in Respiratory Diseases Program at the Research Institute of the McGill University Health Centre, Canada; Department of Pathology, McGill University, Canada
| | - Willem Rijnbout St James
- Meakins-Christie Laboratories, McGill University, Canada; Translational Research in Respiratory Diseases Program at the Research Institute of the McGill University Health Centre, Canada; Department of Pathology, McGill University, Canada
| | - David H Eidelman
- Meakins-Christie Laboratories, McGill University, Canada; Department of Medicine, McGill University, Canada
| | - Carolyn J Baglole
- Meakins-Christie Laboratories, McGill University, Canada; Translational Research in Respiratory Diseases Program at the Research Institute of the McGill University Health Centre, Canada; Department of Pathology, McGill University, Canada; Department of Medicine, McGill University, Canada; Department of Pharmacology and Therapeutics, McGill University, Canada.
| |
Collapse
|
29
|
Diedrich JD, Gonzalez-Pons R, Medeiros HCD, Ensink E, Liby KT, Wellberg EA, Lunt SY, Bernard JJ. Adipocyte-derived kynurenine stimulates malignant transformation of mammary epithelial cells through the aryl hydrocarbon receptor. Biochem Pharmacol 2023; 216:115763. [PMID: 37625554 PMCID: PMC10587895 DOI: 10.1016/j.bcp.2023.115763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/17/2023] [Accepted: 08/21/2023] [Indexed: 08/27/2023]
Abstract
Anti-hormone therapies are not efficacious for reducing the incidence of triple negative breast cancer (TNBC), which lacks both estrogen and progesterone receptors. While the etiology of this aggressive breast cancer subtype is unclear, visceral obesity is a strong risk factor for both pre- and post-menopausal cases. The mechanism by which excessive deposition of visceral adipose tissue (VAT) promotes the malignant transformation of hormone receptor-negative mammary epithelial cells is currently unknown. We developed a novel in vitro system of malignant transformation in which non-tumorigenic human breast epithelial cells (MCF-10A) grow in soft agar when cultured with factors released from VAT. These cells, which acquire the capacity for 3D growth, show elevated aryl hydrocarbon receptor (AhR) protein and AhR target genes, suggesting that AhR activity may drive malignant transformation by VAT. AhR is a ligand-dependent transcription factor that generates biological responses to exogenous carcinogens and to the endogenous tryptophan pathway metabolite, kynurenine. The serum kynurenine to tryptophan ratio has been shown to be elevated in patients with obesity. Herein, we demonstrate that AhR inhibitors or knockdown of AhR in MCF-10A cells prevents VAT-induced malignant transformation. Specifically, VAT-induced transformation is inhibited by Kyn-101, an inhibitor for the endogenous ligand binding site of AhR. Mass spectrometry analysis demonstrates that adipocytes metabolize tryptophan and release kynurenine, which is taken up by MCF-10A cells and activates the AhR to induce CYP1B1 and promote malignant transformation. This novel hormone receptor-independent mechanism of malignant transformation suggests targeting AhR for TNBC prevention in the context of visceral adiposity.
Collapse
Affiliation(s)
- Jonathan D Diedrich
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824 USA
| | - Romina Gonzalez-Pons
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824 USA
| | - Hyllana C D Medeiros
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824 USA
| | - Elliot Ensink
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824 USA
| | - Karen T Liby
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824 USA
| | - Elizabeth A Wellberg
- Department of Pathology, University of Oklahoma Health Sciences Center, Stephenson Cancer Center, Harold Hamm Diabetes Center, Oklahoma City, OK, USA
| | - Sophia Y Lunt
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824 USA; Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI 48824 USA
| | - Jamie J Bernard
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824 USA; Department of Medicine, Michigan State University, East Lansing, MI 48824 USA.
| |
Collapse
|
30
|
Rachdi L, Zhou Z, Berthault C, Lourenço C, Fouque A, Domet T, Armanet M, You S, Peakman M, Mallone R, Scharfmann R. Tryptophan metabolism promotes immune evasion in human pancreatic β cells. EBioMedicine 2023; 95:104740. [PMID: 37536063 PMCID: PMC10412781 DOI: 10.1016/j.ebiom.2023.104740] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 07/18/2023] [Accepted: 07/18/2023] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND To resist the autoimmune attack characteristic of type 1 diabetes, insulin producing pancreatic β cells need to evade T-cell recognition. Such escape mechanisms may be conferred by low HLA class I (HLA-I) expression and upregulation of immune inhibitory molecules such as Programmed cell Death Ligand 1 (PD-L1). METHODS The expression of PD-L1, HLA-I and CXCL10 was evaluated in the human β cell line, ECN90, and in primary human and mouse pancreatic islets. Most genes were determined by real-time RT-PCR, flow cytometry and Western blot. Activator and inhibitor of the AKT signaling were used to modulate PD-L1 induction. Key results were validated by monitoring activity of CD8+ Jurkat T cells presenting β cell specific T-cell receptor and transduced with reporter genes in contact culture with the human β cell line, ECN90. FINDINGS In this study, we identify tryptophan (TRP) as an agonist of PD-L1 induction through the AKT signaling pathway. TRP also synergistically enhanced PD-L1 expression on β cells exposed to interferon-γ. Conversely, interferon-γ-mediated induction of HLA-I and CXCL10 genes was down-regulated upon TRP treatment. Finally, TRP and its derivatives inhibited the activation of islet-reactive CD8+ T cells by β cells. INTERPRETATION Collectively, our findings indicate that TRP could induce immune tolerance to β cells by promoting their immune evasion through HLA-I downregulation and PD-L1 upregulation. FUNDING Dutch Diabetes Research Foundation, DON Foundation, the Laboratoire d'Excellence consortium Revive (ANR-10-LABX-0073), Agence Nationale de la Recherche (ANR-19-CE15-0014-01), Fondation pour la Recherche Médicale (EQ U201903007793-EQU20193007831), Innovative Medicines InitiativeINNODIA and INNODIA HARVEST, Aides aux Jeunes Diabetiques (AJD) and Juvenile Diabetes Research Foundation Ltd (JDRF).
Collapse
Affiliation(s)
- Latif Rachdi
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris 75014, France.
| | - Zhicheng Zhou
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris 75014, France
| | - Claire Berthault
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris 75014, France
| | - Chloe Lourenço
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris 75014, France
| | - Alexis Fouque
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris 75014, France
| | - Thomas Domet
- Assistance Publique Hôpitaux de Paris, Cell Therapy Unit, Saint Louis Hospital, Paris 75010, France
| | - Mathieu Armanet
- Assistance Publique Hôpitaux de Paris, Cell Therapy Unit, Saint Louis Hospital, Paris 75010, France
| | - Sylvaine You
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris 75014, France
| | - Mark Peakman
- Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London SE1 9RT, UK
| | - Roberto Mallone
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris 75014, France; Assistance Publique Hôpitaux de Paris, Service de Diabétologie et Immunologie Clinique, Cochin Hospital, Paris 75014, France
| | - Raphael Scharfmann
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris 75014, France
| |
Collapse
|
31
|
Zhang T, Zhu L, Sun Y, Yang L, Yi S, Zhong W. Novel Insights on 6:6 Perfluoroalkyl Phosphonic Acid-Induced Melanin Synthesis Disorders Leading to Pigmentation in Tadpoles. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:11032-11042. [PMID: 37467139 DOI: 10.1021/acs.est.3c02920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/21/2023]
Abstract
As alternatives to traditional per- and polyfluoroalkyl substances, perfluoroalkyl phosphonic acids (PFPiAs) are widely present in aquatic environments and can potentially harm aquatic organisms. Pigmentation affects the probability of aquatic organisms being preyed on and serves as an important toxic endpoint of development, but little is known about the impacts of PFPiAs on the development of aquatic organisms. In this study, Xenopus laevis embryos were exposed to 6:6 PFPiA (1, 10, and 100 nM) for 14 days. The developed tadpoles exhibited evident pigmentation with increased melanin particle size and density on the skin. Pathological and behavioral experiments revealed that the retinal layers became thinner, reducing the photosensitivity and disturbing the circadian rhythm of the tadpoles. Compared to the control group, the exposed tadpoles showed higher levels but less changes of melanin throughout the light/dark cycle, as well as distinct oxidative damage. Consequently, the expression level of microphthalmia-associated transcription factor (MITF), a key factor inducing melanin synthesis, increased significantly. Molecular docking analysis suggested that 6:6 PFPiA forms strong interactions in the binding pocket of MITF, implying that it could activate MITF directly. The activation of MITF ultimately promotes melanin synthesis, resulting in pigmentation on tadpoles.
Collapse
Affiliation(s)
- Tianxu Zhang
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Lingyan Zhu
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Yumeng Sun
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Liping Yang
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Shujun Yi
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Wenjue Zhong
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| |
Collapse
|
32
|
Sakamoto R, Takada A, Yamakado S, Tsuge H, Ito E, Iwata M. Release from persistent T cell receptor engagement and blockade of aryl hydrocarbon receptor activity enhance IL-6-dependent mouse follicular helper T-like cell differentiation in vitro. PLoS One 2023; 18:e0287746. [PMID: 37352327 PMCID: PMC10289413 DOI: 10.1371/journal.pone.0287746] [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: 09/09/2022] [Accepted: 06/13/2023] [Indexed: 06/25/2023] Open
Abstract
Follicular helper T (Tfh) cells are crucial for humoral immunity. Dysregulation of Tfh cell differentiation can cause infectious, allergic, and autoimmune diseases. To elucidate the molecular mechanisms underlying Tfh cell differentiation, we attempted to establish an in vitro mouse model of Tfh cell differentiation in the absence of other cell types. Various cytokines and cell surface molecules are suggested to contribute to the differentiation. We found that stimulating naïve CD4+ T cells with immobilized antibodies to CD3, ICOS, and LFA-1 in the presence of soluble anti-CD28 antibody, IL-6, and antibodies that block IL-2 signaling for 3 days induced the expression of Bcl6 and Rorc(γt), master regulator genes of Tfh and Th17 cells, respectively. TGF-β significantly enhanced cell proliferation and Bcl6 and Rorc(γt) expression. An additional 2 days of culture without immobilized antibodies selectively downregulated Rorc(γt) expression. These cells produced IL-21 and promoted B cells to produce IgG antibodies. Adding the aryl hydrocarbon receptor (AhR) antagonist CH-223191 to the T cell culture further downregulated Rorc(γt) expression without significantly affecting Bcl6 expression, and upregulated expression of a key Tfh marker, CXCR5. Although their CXCR5 expression levels were still not high, the CH-223191-treated cells showed chemotactic activity towards the CXCR5 ligand CXCL13. On the other hand, AhR agonists upregulated Rorc(γt) expression and downregulated CXCR5 expression. These findings suggest that AhR activity and the duration of T cell receptor stimulation contribute to regulating the balance between Tfh and Th17 cell differentiation. Although this in vitro system needs to be further improved, it may be useful for elucidating the mechanisms of Tfh cell differentiation as well as for screening physiological or pharmacological factors that affect Tfh cell differentiation including CXCR5 expression.
Collapse
Affiliation(s)
- Rei Sakamoto
- Department of Biology, Waseda University, TWIns, Shinjuku, Tokyo, Japan
| | - Ayumi Takada
- Department of Biology, Waseda University, TWIns, Shinjuku, Tokyo, Japan
| | | | - Haruki Tsuge
- Department of Biology, Waseda University, TWIns, Shinjuku, Tokyo, Japan
| | - Etsuro Ito
- Department of Biology, Waseda University, TWIns, Shinjuku, Tokyo, Japan
- Research Organization for Nano and Life Innovation, Waseda University, TWIns, Shinjuku, Tokyo, Japan
| | - Makoto Iwata
- Research Organization for Nano and Life Innovation, Waseda University, TWIns, Shinjuku, Tokyo, Japan
| |
Collapse
|
33
|
Nguyen PT, Nakamura Y, Tran NQV, Ishimaru K, Nguyen TA, Kobayashi Y, Watanabe-Saito F, Okuda T, Nakano N, Nakao A. Ethyl Caffeate Can Inhibit Aryl Hydrocarbon Receptor (AhR) Signaling and AhR-Mediated Potentiation of Mast Cell Activation. Int J Mol Sci 2023; 24:9997. [PMID: 37373144 DOI: 10.3390/ijms24129997] [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: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Ethyl caffeate (EC) is a natural phenolic compound that is present in several medicinal plants used to treat inflammatory disorders. However, its anti-inflammatory mechanisms are not fully understood. Here, we report that EC inhibits aryl hydrocarbon receptor (AhR) signaling and that this is associated with its anti-allergic activity. EC inhibited AhR activation, induced by the AhR ligands FICZ and DHNA in AhR signaling-reporter cells and mouse bone marrow-derived mast cells (BMMCs), as assessed by AhR target gene expressions such as CYP1A1. EC also inhibited the FICZ-induced downregulation of AhR expression and DHNA-induced IL-6 production in BMMCs. Furthermore, the pretreatment of mice with orally administered EC inhibited DHNA-induced CYP1A1 expression in the intestine. Notably, both EC and CH-223191, a well-established AhR antagonist, inhibited IgE-mediated degranulation in BMMCs grown in a cell culture medium containing significant amounts of AhR ligands. Furthermore, oral administration of EC or CH-223191 to mice inhibited the PCA reaction associated with the suppression of constitutive CYP1A1 expression within the skin. Collectively, EC inhibited AhR signaling and AhR-mediated potentiation of mast cell activation due to the intrinsic AhR activity in both the culture medium and normal mouse skin. Given the AhR control of inflammation, these findings suggest a novel mechanism for the anti-inflammatory activity of EC.
Collapse
Affiliation(s)
- Phuc-Tan Nguyen
- Department of Immunology, Faculty of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Yuki Nakamura
- Department of Immunology, Faculty of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Nguyen Quoc Vuong Tran
- Department of Immunology, Faculty of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Kayoko Ishimaru
- Department of Immunology, Faculty of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Thuy-An Nguyen
- Department of Immunology, Faculty of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Yoshiaki Kobayashi
- Department of Immunology, Faculty of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Fumie Watanabe-Saito
- The Institute of Enology and Viticulture, University of Yamanashi, Yamanashi 400-0005, Japan
| | - Tohru Okuda
- The Institute of Enology and Viticulture, University of Yamanashi, Yamanashi 400-0005, Japan
| | - Nobuhiro Nakano
- Atopy Research Center, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| | - Atsuhito Nakao
- Department of Immunology, Faculty of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
- Atopy Research Center, Juntendo University School of Medicine, Tokyo 113-8421, Japan
- Yamanashi GLIA Center, University of Yamanashi, Yamanashi 409-3898, Japan
| |
Collapse
|
34
|
Shi J, Du T, Wang J, Tang C, Lei M, Yu W, Yang Y, Ma Y, Huang P, Chen H, Wang X, Sun J, Wang H, Zhang Y, Luo F, Huang Q, Li B, Lu S, Hu Y, Peng X. Aryl hydrocarbon receptor is a proviral host factor and a candidate pan-SARS-CoV-2 therapeutic target. SCIENCE ADVANCES 2023; 9:eadf0211. [PMID: 37256962 PMCID: PMC10413656 DOI: 10.1126/sciadv.adf0211] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 04/25/2023] [Indexed: 06/02/2023]
Abstract
The emergence of a series of SARS-CoV-2 variants has necessitated the search for broad-spectrum antiviral targets. The aryl hydrocarbon receptor (AhR) senses tryptophan metabolites and is an immune regulator. However, the role of AhR in SARS-CoV-2 infection and whether AhR can be used as the target of antiviral therapy against SARS-CoV-2 and its variants are yet unclear. Here, we show that infection with SARS-CoV-2 activates AhR signaling and facilitates viral replication by interfering with IFN-I-driven antiviral immunity and up-regulating ACE2 receptor expression. The pharmacological AhR blockade or AhR knockout reduces SARS-CoV-2 and its variants' replication in vitro. Drug targeting of AhR with AhR antagonists markedly reduced SARS-CoV-2 and its variants' replication in vivo and ameliorated lung inflammation caused by SARS-CoV-2 infection in hamsters. Overall, AhR was a SARS-CoV-2 proviral host factor and a candidate host-directed broad-spectrum target for antiviral therapy against SARS-CoV-2 and its variants, including Delta and Omicron, and potentially other variants in the future.
Collapse
Affiliation(s)
- Jiandong Shi
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tingfu Du
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junbin Wang
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Cong Tang
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengyue Lei
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenhai Yu
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yun Yang
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Ma
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pu Huang
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongli Chen
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xu Wang
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Sun
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haixuan Wang
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yong Zhang
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fangyu Luo
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qing Huang
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bai Li
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuaiyao Lu
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yunzhang Hu
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaozhong Peng
- National Kunming High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Medical Molecular Biology, Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing China
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing China
| |
Collapse
|
35
|
Ondrová K, Zůvalová I, Vyhlídalová B, Krasulová K, Miková E, Vrzal R, Nádvorník P, Nepal B, Kortagere S, Kopečná M, Kopečný D, Šebela M, Rastinejad F, Pu H, Soural M, Rolfes KM, Haarmann-Stemmann T, Li H, Mani S, Dvořák Z. Monoterpenoid aryl hydrocarbon receptor allosteric antagonists protect against ultraviolet skin damage in female mice. Nat Commun 2023; 14:2728. [PMID: 37169746 PMCID: PMC10174618 DOI: 10.1038/s41467-023-38478-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 05/02/2023] [Indexed: 05/13/2023] Open
Abstract
The human aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that is a pivotal regulator of human physiology and pathophysiology. Allosteric inhibition of AhR was previously thought to be untenable. Here, we identify carvones as noncompetitive, insurmountable antagonists of AhR and characterize the structural and functional consequences of their binding. Carvones do not displace radiolabeled ligands from binding to AhR but instead bind allosterically within the bHLH/PAS-A region of AhR. Carvones do not influence the translocation of ligand-activated AhR into the nucleus but inhibit the heterodimerization of AhR with its canonical partner ARNT and subsequent binding of AhR to the promoter of CYP1A1. As a proof of concept, we demonstrate physiologically relevant Ahr-antagonism by carvones in vivo in female mice. These substances establish the molecular basis for selective targeting of AhR regardless of the type of ligand(s) present and provide opportunities for the treatment of disease processes modified by AhR.
Collapse
Affiliation(s)
- Karolína Ondrová
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | - Iveta Zůvalová
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | - Barbora Vyhlídalová
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | - Kristýna Krasulová
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | - Eva Miková
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | - Radim Vrzal
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | - Petr Nádvorník
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | - Binod Nepal
- Department of Microbiology & Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Sandhya Kortagere
- Department of Microbiology & Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Martina Kopečná
- Department of Experimental Biology, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | - David Kopečný
- Department of Experimental Biology, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | - Marek Šebela
- Department of Biochemistry, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | - Fraydoon Rastinejad
- Target Discovery Institute Nuffield Department of Medicine Research Building Brasenose College University of Oxford, Oxford, UK
| | - Hua Pu
- Target Discovery Institute Nuffield Department of Medicine Research Building Brasenose College University of Oxford, Oxford, UK
| | - Miroslav Soural
- Department of Organic Chemistry, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | | | | | - Hao Li
- Department of Medicine, Oncology, Molecular Pharmacology, and Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Sridhar Mani
- Department of Medicine, Oncology, Molecular Pharmacology, and Genetics, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Zdeněk Dvořák
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Olomouc, Czech Republic.
| |
Collapse
|
36
|
Accioli CDAF, da Silva MS, Santos BAMC, Rodrigues CR. Aryl Hydrocarbon Receptor as a Therapeutical Target of Environmentally Induced Skin Conditions. Mol Pharmacol 2023; 103:255-265. [PMID: 36732021 DOI: 10.1124/molpharm.122.000627] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 01/09/2023] [Accepted: 01/17/2023] [Indexed: 02/04/2023] Open
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor, expressed in several tissues and involved in the response to environmental stressors. Studies have already associated exposure to environmental factors, such as organic air pollutants, products of the skin microbiota, and solar radiation, with the development/worsening of skin conditions, mediated by AhR. On the other hand, recent studies have shown that synthetic and natural compounds are able to modulate the activation of some AhR signaling pathways, minimizing the harmful response of these environmental stressors in the skin. Thus, AhR constitutes a new therapeutic target for the prevention or treatment of skin conditions induced by the skin exposome. Herein, an overview of potential AhR ligands and their biologic effects in environmentally induced skin conditions are presented. The literature survey pointed out divergences in the mechanism of action from a therapeutic perspective. Although most studies point to the benefits of ligand downregulation of AhR signaling, counteracting the toxic effects of environmental factors on the skin, some studies suggest the AhR ligand activation as a therapeutical mechanism for some skin conditions. Furthermore, both agonist and antagonist profiles were identified in the AhR modulation by the synthetic and natural compounds raised. Despite that, this target is still little explored, and further studies are needed to elucidate the molecular mechanisms involved and identify new AhR ligands with therapeutic potential. SIGNIFICANCE STATEMENT: The aryl hydrocarbon receptor (AhR) is involved in different skin physiological and pathological processes, including toxic mechanisms of environmental factors. Synthetic and natural AhR ligands have demonstrated therapeutic potential for skin conditions induced by these agents. Thus, a comprehensive understanding of the skin toxicity mechanisms involving the AhR, as well as the use of AhR modulators from a therapeutic perspective, provides an alternative approach to the development of new treatments for skin disorders induced by the exposome.
Collapse
Affiliation(s)
- Caroline de Almeida Freitas Accioli
- Laboratório de Planejamento Farmacêutico e Simulação Computacional (LaPFarSC) (C.d.A.F.A., B.A.M.C.S.) and Laboratório de Modelagem Molecular & QSAR-3D (ModMolQSAR), Departamento de Fármacos e Medicamentos (DEFARMED) (C.R.R.), Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil and MEDCINVITRO, São Paulo, Brazil (M.S.d.S.)
| | - Michelle Sabrina da Silva
- Laboratório de Planejamento Farmacêutico e Simulação Computacional (LaPFarSC) (C.d.A.F.A., B.A.M.C.S.) and Laboratório de Modelagem Molecular & QSAR-3D (ModMolQSAR), Departamento de Fármacos e Medicamentos (DEFARMED) (C.R.R.), Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil and MEDCINVITRO, São Paulo, Brazil (M.S.d.S.)
| | - Bianca Aloise Maneira Corrêa Santos
- Laboratório de Planejamento Farmacêutico e Simulação Computacional (LaPFarSC) (C.d.A.F.A., B.A.M.C.S.) and Laboratório de Modelagem Molecular & QSAR-3D (ModMolQSAR), Departamento de Fármacos e Medicamentos (DEFARMED) (C.R.R.), Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil and MEDCINVITRO, São Paulo, Brazil (M.S.d.S.)
| | - Carlos Rangel Rodrigues
- Laboratório de Planejamento Farmacêutico e Simulação Computacional (LaPFarSC) (C.d.A.F.A., B.A.M.C.S.) and Laboratório de Modelagem Molecular & QSAR-3D (ModMolQSAR), Departamento de Fármacos e Medicamentos (DEFARMED) (C.R.R.), Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil and MEDCINVITRO, São Paulo, Brazil (M.S.d.S.)
| |
Collapse
|
37
|
Elson DJ, Kolluri SK. Tumor-Suppressive Functions of the Aryl Hydrocarbon Receptor (AhR) and AhR as a Therapeutic Target in Cancer. BIOLOGY 2023; 12:526. [PMID: 37106727 PMCID: PMC10135996 DOI: 10.3390/biology12040526] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/25/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor involved in regulating a wide range of biological responses. A diverse array of xenobiotics and endogenous small molecules bind to the receptor and drive unique phenotypic responses. Due in part to its role in mediating toxic responses to environmental pollutants, AhR activation has not been traditionally viewed as a viable therapeutic approach. Nonetheless, the expression and activation of AhR can inhibit the proliferation, migration, and survival of cancer cells, and many clinically approved drugs transcriptionally activate AhR. Identification of novel select modulators of AhR-regulated transcription that promote tumor suppression is an active area of investigation. The development of AhR-targeted anticancer agents requires a thorough understanding of the molecular mechanisms driving tumor suppression. Here, we summarized the tumor-suppressive mechanisms regulated by AhR with an emphasis on the endogenous functions of the receptor in opposing carcinogenesis. In multiple different cancer models, the deletion of AhR promotes increased tumorigenesis, but a precise understanding of the molecular cues and the genetic targets of AhR involved in this process is lacking. The intent of this review was to synthesize the evidence supporting AhR-dependent tumor suppression and distill insights for development of AhR-targeted cancer therapeutics.
Collapse
Affiliation(s)
- Daniel J. Elson
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA
| | - Siva K. Kolluri
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
| |
Collapse
|
38
|
Mi P, Li N, Ai K, Li L, Yuan D. AhR-mediated lipid peroxidation contributes to TCDD-induced cardiac defects in zebrafish. CHEMOSPHERE 2023; 317:137942. [PMID: 36702031 DOI: 10.1016/j.chemosphere.2023.137942] [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: 10/04/2022] [Revised: 01/20/2023] [Accepted: 01/21/2023] [Indexed: 06/18/2023]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD), a persistent environmental contaminant that activates the aryl hydrocarbon receptor (AhR) pathway, has been reported to cause cardiac damage. However, the mechanism underlying AhR-induced cardiac defects in response to TCDD exposure remains unclear. In this study, we characterized the impacts of TCDD exposure on heart morphology and cardiac function in zebrafish. TCDD exposure in the early developmental stage of zebrafish embryos led to morphological heart malformation and pericardial edema, concomitant with reduced cardiac function. These cardiac defects were attenuated by inhibiting AhR activity with CH223191. Transcriptome profiling showed that, along with an upregulation of the AhR signaling pathway by TCDD treatment, the expression of pro-ferroptotic genes was upregulated, while that of genes implicated in glutathione metabolism were downregulated. Moreover, lipid peroxidation, as indicated by malonaldehyde (MDA) production, was increased in TCDD-exposed cardiac tissue. Accordingly, inhibiting lipid peroxidation with liproxstatin-1 reversed the adverse cardiac effects induced by TCDD treatment. Taken together, our findings demonstrate that AhR-mediated lipid peroxidation contributes to cardiac defects in the early developmental stage in zebrafish embryos exposed to TCDD.
Collapse
Affiliation(s)
- Ping Mi
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China; Key Laboratory of Experimental Teratology, Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
| | - Na Li
- Department of Histology and Embryology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China; Key Laboratory of Experimental Teratology, Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
| | - Kang Ai
- Department of Histology and Embryology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China; Key Laboratory of Experimental Teratology, Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
| | - Lei Li
- Department of Histology and Embryology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China; Key Laboratory of Experimental Teratology, Ministry of Education, Shandong University, Jinan, Shandong, 250012, China.
| | - Detian Yuan
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
39
|
Tanabe P, Schlenk D. Role of Aryl Hydrocarbon Receptor and Oxidative Stress in the Regioselective Toxicities of Hydroxychrysenes in Embryonic Japanese Medaka (Oryzias latipes). ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2023; 42:698-706. [PMID: 36636887 DOI: 10.1002/etc.5560] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/16/2022] [Accepted: 01/05/2023] [Indexed: 06/17/2023]
Abstract
Oxygenated polycyclic aromatic hydrocarbons (oxy-PAHs) are environmental contaminants that can be created through oxidation of parent PAHs. Previous studies have found that 2-hydroxychrysene (2-OHCHR) caused anemia in embryonic Japanese medaka whereas 6-hydroxychrysene (6-OHCHR) did not, an example of regioselective toxicity. Anemia was prevented by cytochrome P450 (CYP) inhibition, which reduced the formation of the potential oxidatively active metabolite, 1,2-catechol, from 2-OHCHR. 2-OHCHR has also been found to be a four-fold more potent aryl hydrocarbon receptor (AhR) agonist compared with 6-OHCHR. These findings led us to hypothesize that AhR activation and/or oxidative stress play an important role in 2-OHCHR toxicity. Although treatments with the AhR agonists polychlorinated biphenyl (PCB)126 and 2-methoxychrysene (2-MeOCHR) did not cause significant toxicity, pretreatments with the AhR antagonist, CH-223191, reduced anemia by 97.2 ± 0.84% and mortality by 96.6 ± 0.69%. Aryl hydrocarbon receptor inhibition by the antagonist was confirmed by significant reductions (91.0 ± 9.94%) in induced ethoxyresorufin-O-deethylase activity. Thiobarbituric acid reactive substances concentrations were 32.9 ± 3.56% higher (p < 0.05) in 2-OHCHR treatments at 100 hours postfertilization compared with controls. Staining 2-OHCHR-treated embryos with the reactive oxygen species (ROS) scavenger 2',7'-dichlorofluorescin diacetate revealed 32.6 ± 2.69% of 2-OHCHR-treated embryos exhibiting high concentrations of ROS in caudal tissues, which is a site for embryonic hematopoiesis in medaka. Pretreatment with antioxidants, N-acetylcysteine (NAC) or vitamin E (Vit E) significantly reduced 2-OHCHR-induced anemia (NAC: 80.7 ± 1.12% and Vit E: 99.1 ± 0.43%) and mortality (NAC: 67.1 ± 1.69% and Vit E: 98.9 ± 0.66%). These results indicate that AhR may mediate 2-OHCHR toxicity through canonical signaling by up-regulating CYP1, enhancing the formation of reactive metabolites of 2-OHCHR that generate ROS within caudal hematopoietic tissues, potentially disrupting hematopoiesis, leading to anemia and subsequent mortality. Environ Toxicol Chem 2023;42:698-706. © 2023 The Authors. Environmental Toxicology and Chemistry published by Wiley Periodicals LLC on behalf of SETAC.
Collapse
Affiliation(s)
- Philip Tanabe
- Environmental Toxicology Graduate Program, University of California, Riverside, California, USA
- Department of Environmental Sciences, University of California, Riverside, California, USA
| | - Daniel Schlenk
- Department of Environmental Sciences, University of California, Riverside, California, USA
| |
Collapse
|
40
|
Lee KH, Rim DE, Lee JH, Jeong SW. Role of ATP5G3 in sodium nitroprusside-induced cell death in cervical carcinoma cells. J Biochem Mol Toxicol 2023; 37:e23267. [PMID: 36524533 DOI: 10.1002/jbt.23267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 09/20/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022]
Abstract
We identified a gene, subunit C3 (ATP5G3) of mitochondrial ATP synthase, that displayed changes in gene expression under oxidative stress. We examined the role of ATP5G3 and its molecular mechanisms in sodium nitroprusside (SNP)-induced cell death using ATP5G3 small interfering RNA (siATP5G3)-transfected HeLa cells. A significant increase in cytotoxicity was observed in the transfected cells treated with SNP, which suggests a protective role of ATP5G3 in SNP-induced cytotoxicity in the cells. The transfected cells treated with photodegraded SNP showed equal cytotoxicity to SNP, and pretreatment with deferoxamine (DFO) completely inhibited this cytotoxicity. Further, cytotoxicity was significantly inhibited by pretreatment with a p38 inhibitor and was accentuated by the p38 activator in cells. Pretreatment with the Bcl-xL inhibitor also significantly accentuated cytotoxicity. The increase in p38 phosphorylation was significantly higher in siATP5G3-transfected cells treated with SNP in immunoblotting, which was inhibited by pretreatment with DFO. The increase in cytotoxicity with siATP5G3 transfection was completely blocked by cotransfection with sip38, and the blocking effect disappeared by cotransfection with additional siBcl-xL, which suggests that the protective role of ATP5G3 is mediated by Bcl-xL via the inhibition of p38 activity. Cytotoxicity was completely blocked by the cotransfection of siATP5G3 with siBax. No change in apoptotic parameters was observed during cytotoxicity. However, pretreatment with lysosomal inhibitors significantly inhibited cytotoxicity and increased p62 protein levels. These findings suggest that ATP5G3 plays a protective role in autophagic cell death/lysosome-associated cell death induced by SNP via the sequential signaling of ROS/p38/Bcl-xL/Bax in HeLa cells.
Collapse
Affiliation(s)
- Kyung Hye Lee
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Do Eun Rim
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jeong-Hwa Lee
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seong-Whan Jeong
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
41
|
Liang S, Zhang Y, Bo H, Duan W, Zhong L. Insights into the toxicities of UV-328, UV-329, UV-P in HepG2 cells and their roles in AHR-mediated pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 250:114478. [PMID: 36586167 DOI: 10.1016/j.ecoenv.2022.114478] [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: 07/20/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 06/17/2023]
Abstract
The widespread high concentrations of benzotriazole ultraviolet stabilizers (BUVSs) in many biotic and abiotic samples have raised urgent concerns of their adverse effects on environmental and human health. In this study, we investigated the toxicity of three typical BUVSs (UV-328, UV-329, UV-P) with HepG2 cells in vitro. Results indicated that the three BUVSs showed weak cytotoxicity in HepG2 cells at concentrations lower than 50 μM. Transcriptional analysis indicated that the toxic effects of the three chemicals followed the order of UV-P > UV-329 > UV-328. UV-P and UV-329 may act as potential environmental diabetogens by significantly enriching several diabetic related items in both GO and KEGG analysis. Moreover, UV-P and UV-329 significantly upregulated the expression of AHR target genes (CYP1A1, CYP1A2, UGT1A1, etc.), and increased the ethoxyresorufin-O-deethylase (EROD) activity and exhibited agonistic activity toward AHR in the XRE-mediated luciferase reporter gene assay. Molecular docking assay also indicated that UV-329 and UV-P had higher binding affinities to AHR-LBD than UV-328. In brief, our findings indicated that UV-P and UV-329 were potential agonist of AHR ligand, and may exert more toxicity than UV-328 in inducing liver toxicity.
Collapse
Affiliation(s)
- Shengxian Liang
- Institute of Life Sciences and Green Development, College of Life Sciences, Hebei University, Baoding 071000, China.
| | - Yue Zhang
- Institute of Life Sciences and Green Development, College of Life Sciences, Hebei University, Baoding 071000, China
| | - Haimei Bo
- Institute of Life Sciences and Green Development, College of Life Sciences, Hebei University, Baoding 071000, China
| | - Wenzhao Duan
- Institute of Life Sciences and Green Development, College of Life Sciences, Hebei University, Baoding 071000, China
| | - Li Zhong
- Institute of Life Sciences and Green Development, College of Life Sciences, Hebei University, Baoding 071000, China; Western University of Health Sciences, Pomona, CA 91766, USA.
| |
Collapse
|
42
|
Zhang S, Funahashi Y, Tanaka S, Okubo T, Thapa J, Nakamura S, Higashi H, Yamaguchi H. Chlamydia trachomatis relies on the scavenger role of aryl hydrocarbon receptor with detyrosinated tubulin for its intracellular growth, but this is impaired by excess indole. Microbes Infect 2023; 25:105097. [PMID: 36608767 DOI: 10.1016/j.micinf.2022.105097] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/29/2022] [Accepted: 12/29/2022] [Indexed: 01/05/2023]
Abstract
Although IFN-γ depletes tryptophan (Trp) as a defense against intracellular Chlamydia trachomatis (Ct) infected to hypoxic vagina, the presence of indole, a precursor of Trp, enables Ct to infect IFN-γ-exposed culture cells. Meanwhile, Trp-derived indole derivatives interact the aryl hydrocarbon receptor (AhR), which is a ligand-dependent transcription factor involved in the cellular homeostasis with tubulin dynamics. Here, the amounts of IFN-γ and indole in cervical swabs with known Ct infection status were measured, and Ct growth in the presence of indole was determined from the perspective of the AhR axis under hypoxia. A positive correlation between the amounts of IFN-γ and indole was found, and both of these amounts were lower in Ct-positive swabs than in Ct-negative ones. Indole as well as other AhR ligands inhibited Ct growth, especially under normoxia. Ct prompted the expression of detyrosinated tubulin (dTTub), but indole inhibited it. Indole did not stimulate the translocation of AhR to nucleus, and it blocked AhR activation in AhR-reporter cells. Ct growth was reduced more effectively under normoxia in AhR-knockdown cells, an effect that was enhanced by indole, which in turn diminished dTTub. Thus, Ct growth relies on the scavenger role of cytosolic AhR responsible for promoting dTTub expression.
Collapse
Affiliation(s)
- Saicheng Zhang
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, North-12, West-5, Kita-ku, Sapporo 060-0812, Japan.
| | - Yuki Funahashi
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, North-12, West-5, Kita-ku, Sapporo 060-0812, Japan.
| | - Satoho Tanaka
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, North-12, West-5, Kita-ku, Sapporo 060-0812, Japan.
| | - Torahiko Okubo
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, North-12, West-5, Kita-ku, Sapporo 060-0812, Japan.
| | - Jeewan Thapa
- Division of Bioresources, International Institute for Zoonosis Control, Hokkaido University, North-20, West-10, Kita-ku, Sapporo 001-0020, Japan.
| | - Shinji Nakamura
- Division of Biomedical Imaging Research, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; Division of Ultrastructural Research, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.
| | - Hideaki Higashi
- Division of Infection and Immunity, International Institute for Zoonosis Control, Hokkaido University, North-20, West-10, Kita-ku, Sapporo 001-0020, Japan.
| | - Hiroyuki Yamaguchi
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, North-12, West-5, Kita-ku, Sapporo 060-0812, Japan.
| |
Collapse
|
43
|
In-depth analysis of the interactions of various aryl hydrocarbon receptor ligands from a computational perspective. J Mol Graph Model 2023; 118:108339. [PMID: 36183684 DOI: 10.1016/j.jmgm.2022.108339] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 09/01/2022] [Accepted: 09/17/2022] [Indexed: 11/21/2022]
Abstract
Aryl hydrocarbon receptor (AhR) is a ligand-dependent transcription factor that acts as a machinery that controls the expression of many genes, including cytochrome P450 CYP1A1, CYP1A2 and CYP1B1. It plays a principal role in numerous biological and toxicological functions, making it a promising target for developing therapeutic agents. Several novel small molecules targeting the AhR signaling pathway are currently under investigation as antitumor agents. Some have already advanced into clinical trials in patients with various tumors. Activation of AhR by diverse chemicals either endogenous or exogenous is initiated by the binding of these ligands to the PAS-B domain, which modulates AhR functions. There is, however, limited information about how various ligands interact with the PAS-B domain for activating or inhibiting the AhR. To better understand the mode of action of AhR agonists/antagonists. The current work proposes a combination of several computational tools to build dynamical models for the PAS-B domain bound to different ligands in mouse and human. Our findings reveal the essential roles of specific PAS-B residues (e.g., S365, V381& Q383), which mediate the AhR ligand-binding process. Our results also explain how these residues regulate the promiscuity of AhR in accommodating various chemicals in its binding PAS-B ligand-binding pocket.
Collapse
|
44
|
Feng R, Lu M, Yin C, Xu K, Liu L, Xu P. Identification of candidate genes and pathways associated with juvenile idiopathic arthritis by integrative transcriptome-wide association studies and mRNA expression profiles. Arthritis Res Ther 2023; 25:19. [PMID: 36755318 PMCID: PMC9906884 DOI: 10.1186/s13075-023-03003-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 01/31/2023] [Indexed: 02/10/2023] Open
Abstract
AIM Juvenile idiopathic arthritis (JIA) is the most common chronic rheumatic disease of childhood, with genetic susceptibility and pathological processes such as autoimmunity and autoinflammation, but its pathogenesis is unclear. We conducted a transcriptome-wide association study (TWAS) using expression interpolation from a large-scale genome-wide association study (GWAS) dataset to identify genes, biological pathways, and environmental chemicals associated with JIA. METHODS We obtained published GWAS data on JIA for TWAS and used mRNA expression profiling to validate the genes identified by TWAS. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed. A protein-protein interaction (PPI) network was generated, and central genes were obtained using Molecular Complex Detection (MCODE). Finally, chemical gene expression datasets were obtained from the Comparative Toxicogenomics database for chemical genome enrichment analysis. RESULTS TWAS identified 1481 genes associated with JIA, and 154 differentially expressed genes were identified based on mRNA expression profiles. After comparing the results of TWAS and mRNA expression profiles, we obtained eight overlapping genes. GO and KEGG enrichment analyses of the genes identified by TWAS yielded 163 pathways, and PPI network analysis as well as MCODE resolution identified a total of eight clusters. Through chemical gene set enrichment analysis, 287 environmental chemicals associated with JIA were identified. CONCLUSION By integrating TWAS and mRNA expression profiles, genes, biological pathways, and environmental chemicals associated with JIA were identified. Our findings provide new insights into the pathogenesis of JIA, including candidate genetic and environmental factors contributing to its onset and progression.
Collapse
Affiliation(s)
- Ruoyang Feng
- grid.452452.00000 0004 1757 9282Department of Joint Surgery, HongHui Hospital, Xian Jiaotong University, Xi’an, 710054 Shanxi China
| | - Mengnan Lu
- grid.452672.00000 0004 1757 5804Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi China
| | - Chunyan Yin
- grid.452672.00000 0004 1757 5804Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi China
| | - Ke Xu
- grid.452452.00000 0004 1757 9282Department of Joint Surgery, HongHui Hospital, Xian Jiaotong University, Xi’an, 710054 Shanxi China
| | - Lin Liu
- grid.452452.00000 0004 1757 9282Department of Joint Surgery, HongHui Hospital, Xian Jiaotong University, Xi’an, 710054 Shanxi China
| | - Peng Xu
- Department of Joint Surgery, HongHui Hospital, Xian Jiaotong University, Xi'an, 710054, Shanxi, China.
| |
Collapse
|
45
|
Hattori N, Nakagawa T, Yoneda M, Hayashida H, Nakagawa K, Yamamoto K, Htun MW, Shibata Y, Koji T, Ito T. Compounds in cigarette smoke induce EGR1 expression via the AHR, resulting in apoptosis and COPD. J Biochem 2022; 172:365-376. [PMID: 36200927 DOI: 10.1093/jb/mvac077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 09/13/2022] [Indexed: 12/13/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a major cause of mortality worldwide, and pulmonary epithelial cell apoptosis is regarded as one of the most important factors in its pathogenesis. Here, we examined the molecular mechanisms of apoptosis caused by cigarette smoke (CS). In the normal bronchial epithelium cell line BEAS-2B, a CS extract markedly induced apoptosis together with transient early growth response 1 (EGR1) protein expression, which is activated over time via the aryl hydrocarbon receptor (AHR). The CS extract-induced apoptosis decreased cell count of BEAS-2B cells and was significantly reversed by knockdown of either EGR1 or AHR. In vivo, the CS extract caused alveolar wall destruction, mimicking COPD, 1 week after intrathoracic injection. Bronchoalveolar lavage fluid (BALF) from the CS extract-treated mice contained massive numbers of apoptotic epithelial cells. Furthermore, it was found that aminoanthracene induced EGR1 expression and cell apoptosis. By contrast, the AHR antagonist stemregenin 1 (SR1) restored apoptosis upon CS treatment. These results suggest that aryl hydrocarbons, such as aminoanthracene, induce EGR1 expression via the AHR, resulting in cell apoptosis and that this can be prevented by administration of an antagonist of AHR.
Collapse
Affiliation(s)
- Naoko Hattori
- Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8523, Japan.,Department of Biochemistry, Nagasaki University School of Medicine, Nagasaki 852-8523, Japan.,Department of Dermatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8523, Japan
| | - Takeya Nakagawa
- Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8523, Japan.,Department of Biochemistry, Nagasaki University School of Medicine, Nagasaki 852-8523, Japan
| | - Mitsuhiro Yoneda
- Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8523, Japan.,Department of Biochemistry, Nagasaki University School of Medicine, Nagasaki 852-8523, Japan
| | - Hiromi Hayashida
- Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8523, Japan.,Department of Biochemistry, Nagasaki University School of Medicine, Nagasaki 852-8523, Japan
| | - Kaori Nakagawa
- Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8523, Japan.,Department of Biochemistry, Nagasaki University School of Medicine, Nagasaki 852-8523, Japan
| | - Kazuo Yamamoto
- Biomedical Research Support Center, Nagasaki University School of Medicine, Nagasaki 852-8523, Japan
| | - Myo Win Htun
- Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8523, Japan.,Department of Histology and Cell Biology, Nagasaki University School of Medicine, Nagasaki 852-8523, Japan
| | - Yasuaki Shibata
- Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8523, Japan.,Department of Dermatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8523, Japan
| | - Takehiko Koji
- Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8523, Japan.,Department of Histology and Cell Biology, Nagasaki University School of Medicine, Nagasaki 852-8523, Japan
| | - Takashi Ito
- Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8523, Japan.,Department of Biochemistry, Nagasaki University School of Medicine, Nagasaki 852-8523, Japan
| |
Collapse
|
46
|
Cerracchio C, Serra F, Amoroso MG, Fiorito F. Canine Coronavirus Activates Aryl Hydrocarbon Receptor during In Vitro Infection. Viruses 2022; 14:v14112437. [PMID: 36366535 PMCID: PMC9692492 DOI: 10.3390/v14112437] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/20/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that interacts with substrates, including microbial metabolites. Recent advances reveal that AhR is involved in the host response to coronaviruses (CoVs) infection. Particularly, AhR antagonists decrease the expression of angiotensin-converting enzyme 2 (ACE2) via AhR up-regulation, resulting in suppression of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection in mammalian cells. Herein, we report that AhR is expressed in canine fibrosarcoma (A72) cells, where it is considerably activated by infection with genotype II of canine coronavirus (CCoV-II). The pharmacological inhibition of AhR, by CH223191, suppressed cell death signs and increased cell viability. Furthermore, the AhR antagonist induced a meaningful decline in virus yield, accompanied by the inhibition of the expression of viral nuclear protein (NP). Fascinatingly, during CCoV infection, a novel co-expression of NP and AhR expression was found. Taken together, our preliminary findings show that infection with CCoV activates AhR, and pharmacologic AhR inhibition reduces CCoV replication, identifying AhR as a possible candidate target for CCoV antiviral therapy.
Collapse
Affiliation(s)
- Claudia Cerracchio
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy
| | - Francesco Serra
- Department of Animal Health, Unit of Virology, Istituto Zooprofilattico del Mezzogiorno, 80055 Portici, Naples, Italy
| | - Maria Grazia Amoroso
- Department of Animal Health, Unit of Virology, Istituto Zooprofilattico del Mezzogiorno, 80055 Portici, Naples, Italy
| | - Filomena Fiorito
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy
- Correspondence: (F.F.); Tel.: +39-0812536180
| |
Collapse
|
47
|
Cao W, Lu J, Li L, Qiu C, Qin X, Wang T, Li S, Zhang J, Xu J. Activation of the Aryl Hydrocarbon Receptor Ameliorates Acute Rejection of Rat Liver Transplantation by Regulating Treg Proliferation and PD-1 Expression. Transplantation 2022; 106:2172-2181. [PMID: 35706097 DOI: 10.1097/tp.0000000000004205] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Aryl hydrocarbon receptor (AhR) plays important roles in modulating immune responses. However, the role of AhR in rat liver transplantation (LT) has not been explored. METHODS Safety and side effects of N -(3,4-dimethoxycinnamonyl) anthranilic acid (3,4-DAA) and 2-methyl-2H-pyrazole-3-carboxylic acid amide (CH223191) were evaluated. We used optimal doses of 2 drugs, 3,4-DAA, a drug used for mediating AhR activation, and CH223191, antagonist of AhR (3,4-DAA, CH223191, and 3,4-DAA + CH223191), intraperitoneally administered to recipients daily to investigate the role of AhR in the rat LT model. The recipient livers were used to observe the pathological changes, the cells infiltrating the graft, and changes of AhR and programmed death-1 (PD-1) by Western blot, real-time polymerase chain reaction, and immunofluorescence assays. The contents of Foxp3 + and PD-1 + T cells in the recipient spleen and peripheral blood mononuclear cells were evaluated by flow cytometry. In vitro, after isolating CD4 + T cells, they were treated with different AhR ligands to observe the differentiation direction and PD-1 expression level. RESULTS The activation of AhR by 3,4-DAA prolonged survival time and ameliorated graft rejection, which were associated with increased expression of AhR and PD-1 in the livers and increased Foxp3 + T cells and PD-1 + T cells in recipient spleens, livers, and peripheral blood mononuclear cells. In vitro, primary T cells incubated with 3,4-DAA mediated increased proportion of Treg and PD-1 + T cells. However, the suppression of AhR with CH223191 reverses these effects, both in the LT model and in vitro. CONCLUSIONS Our results indicated that AhR activation might reduce the occurrence of rat acute rejection by increasing the proportion of Treg and the expression of PD-1.
Collapse
Affiliation(s)
- Wanyue Cao
- Department of Hepatobiliary Surgery and Liver Transplantation, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Lu
- Department of Hepatobiliary Surgery and Liver Transplantation, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Li
- Department of Hepatobiliary Surgery and Liver Transplantation, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Qiu
- Department of General Surgery, Center of Gallbladder Disease, Shanghai East Hospital, Institute of Gallstone Disease, School of Medicine, Tongji University, Shanghai, China
| | - Xuebin Qin
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA
- Department of Neuroscience, Temple University Lewis Katz School of Medicine, Philadelphia, PA
| | - Tao Wang
- Department of Hepatobiliary Surgery and Liver Transplantation, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shanbao Li
- Department of Hepatobiliary Surgery and Liver Transplantation, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinyan Zhang
- Department of Hepatobiliary Surgery and Liver Transplantation, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junming Xu
- Department of Hepatobiliary Surgery and Liver Transplantation, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
48
|
Sakakibara Y, Kojima A, Asai Y, Nadai M, Katoh M. Changes in uridine 5'-diphospho-glucuronosyltransferase 1A6 expression by histone deacetylase inhibitor valproic acid. Biopharm Drug Dispos 2022; 43:175-182. [PMID: 36000181 DOI: 10.1002/bdd.2328] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 08/10/2022] [Accepted: 08/18/2022] [Indexed: 11/09/2022]
Abstract
Valproic acid (VPA) is well-known as a histone deacetylase (HDAC) inhibitor. It has been reported that HDAC inhibitors enhance basal and aryl hydrocarbon receptor (AhR) ligand-induced aryl hydrocarbon receptor-responsive gene expression. Other studies suggested that HDAC inhibition might significantly activate the NF-E2-related factor-2 (Nrf2). Moreover, VPA activates mitogen-activated protein kinases (MAPKs). MAPK pathways regulate Nrf2 transactivation domain activity. Uridine 5'-diphospho-glucuronosyltransferase (UGT) 1A6 is one of the important isoforms to affect drug pharmacokinetics. UGT1A6 gene is regulated transcriptionally by AhR and Nrf2. The present study aimed to investigate whether UGT1A6 expression was changed by VPA and to elucidate the mechanism of the alteration. Following VPA treatment for 72 h in Caco-2 cells, UGT1A6 mRNA was increased by 7.9-fold. Moreover, UGT1A6 mRNA was increased by other HDAC inhibitors, suggesting that HDAC inhibition caused the UGT1A6 mRNA induction. AhR and Nrf2 proteins in the nucleus of Caco-2 cells were increased by 1.5- and 1.7-fold, respectively, following the VPA treatment. However, VPA treatment did not activate the extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) pathways in Caco-2 cells. In conclusion, we observed that VPA induced UGT1A6 mRNA expression via AhR and Nrf2 pathways, but not via the ERK or JNK pathways.
Collapse
Affiliation(s)
| | - Ayaka Kojima
- Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Yuki Asai
- Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | | | - Miki Katoh
- Faculty of Pharmacy, Meijo University, Nagoya, Japan
| |
Collapse
|
49
|
McGovern K, Castro AC, Cavanaugh J, Coma S, Walsh M, Tchaicha J, Syed S, Natarajan P, Manfredi M, Zhang XM, Ecsedy J. Discovery and Characterization of a Novel Aryl Hydrocarbon Receptor Inhibitor, IK-175, and Its Inhibitory Activity on Tumor Immune Suppression. Mol Cancer Ther 2022; 21:1261-1272. [PMID: 35666806 DOI: 10.1158/1535-7163.mct-21-0984] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 04/01/2022] [Accepted: 05/27/2022] [Indexed: 12/13/2022]
Abstract
Aryl hydrocarbon receptor (AHR) is a transcription factor that regulates the activity of multiple innate and adaptive immune cells subsequent to binding to numerous endogenous and exogenous ligands. For example, AHR is activated by the metabolite kynurenine, which is secreted into the tumor microenvironment by cancer cells leading to broad immunosuppression. Therefore, AHR inhibition provides a novel and ideal approach to stimulate immune-mediated recognition and subsequent eradication of tumor cells. We report here the discovery and characterization of IK-175, a novel, potent and selective AHR antagonist with favorable ADME and pharmacokinetic profiles in preclinical species. IK-175 inhibits AHR activity in experimental systems derived from multiple species including mouse, rat, monkey, and humans. In human primary immune cells, IK-175 decreased AHR target gene expression and anti-inflammatory cytokine release and increased proinflammatory cytokine release. Moreover, IK-175 led to a decrease in suppressive IL17A-, IL-22+ expressing T cells in a Th17 differentiation assay. IK-175 dose dependently blocks ligand-stimulated AHR activation of Cyp1a1 transcription in mouse liver and spleen, demonstrating on-target in vivo activity. IK-175 increases proinflammatory phenotype of the tumor microenvironment in mouse syngeneic tumors and in adjacent tumor-draining lymph nodes. As a monotherapy and combined with an anti-PD-1 antibody, IK-175 demonstrates antitumor activity in syngeneic mouse models of colorectal cancer and melanoma. IK-175 also demonstrates antitumor activity combined with liposomal doxorubicin in syngeneic mouse tumors. These studies provide rationale for targeting AHR in patients with cancer. IK-175 is being evaluated in a phase I clinical trial in patients with advanced solid tumors.
Collapse
|
50
|
Peng G, Tsukamoto S, Ikutama R, Le Thanh Nguyen H, Umehara Y, Trujillo-Paez JV, Yue H, Takahashi M, Ogawa T, Kishi R, Tominaga M, Takamori K, Kitaura J, Kageyama S, Komatsu M, Okumura K, Ogawa H, Ikeda S, Niyonsaba F. Human-β-defensin-3 attenuates atopic dermatitis-like inflammation through autophagy activation and the aryl hydrocarbon receptor signaling pathway. J Clin Invest 2022; 132:156501. [PMID: 35834333 PMCID: PMC9435650 DOI: 10.1172/jci156501] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 07/12/2022] [Indexed: 01/18/2023] Open
Abstract
Human β-defensin-3 (hBD-3) exhibits antimicrobial and immunomodulatory activities; however, its contribution to autophagy regulation remains unclear, and the role of autophagy in the regulation of the epidermal barrier in atopic dermatitis (AD) is poorly understood. Here, keratinocyte autophagy was restrained in the skin lesions of patients with AD and murine models of AD. Interestingly, hBD-3 alleviated the IL-4– and IL-13–mediated impairment of the tight junction (TJ) barrier through keratinocyte autophagy activation, which involved aryl hydrocarbon receptor (AhR) signaling. While autophagy deficiency impaired the epidermal barrier and exacerbated inflammation, hBD-3 attenuated skin inflammation and enhanced the TJ barrier in AD. Importantly, hBD-3–mediated improvement of the TJ barrier was abolished in autophagy-deficient AD mice and in AhR-suppressed AD mice, suggesting a role for hBD-3–mediated autophagy in the regulation of the epidermal barrier and inflammation in AD. Thus, autophagy contributes to the pathogenesis of AD, and hBD-3 could be used for therapeutic purposes.
Collapse
Affiliation(s)
- Ge Peng
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Saya Tsukamoto
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Risa Ikutama
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hai Le Thanh Nguyen
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoshie Umehara
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Juan V Trujillo-Paez
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hainan Yue
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Miho Takahashi
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Takasuke Ogawa
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ryoma Kishi
- Institute for Environmental and Gender Specific Medicine, Juntendo University Graduate School of Medicine, Chiba, Japan
| | - Mitsutoshi Tominaga
- Institute for Environmental and Gender Specific Medicine, Juntendo University Graduate School of Medicine, Chiba, Japan
| | - Kenji Takamori
- Institute for Environmental and Gender Specific Medicine, Juntendo University Graduate School of Medicine, Chiba, Japan
| | - Jiro Kitaura
- Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shun Kageyama
- Department of Physiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masaaki Komatsu
- Department of Physiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ko Okumura
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hideoki Ogawa
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shigaku Ikeda
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - François Niyonsaba
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|