1
|
Abduh MS, Alwassil OI, Aldaqal SM, Alfwuaires MA, Farhan M, Hanieh H. A pyrazolopyridine as a novel AhR signaling activator with anti-breast cancer properties in vitro and in vivo. Biochem Pharmacol 2024; 222:116079. [PMID: 38402910 DOI: 10.1016/j.bcp.2024.116079] [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: 10/06/2023] [Revised: 01/29/2024] [Accepted: 02/22/2024] [Indexed: 02/27/2024]
Abstract
Breast cancer is one of the main causes of malignancy-related deaths globally and has a significant impact on women's quality of life. Despite significant therapeutic advances, there is a medical need for targeted therapies in breast cancer. Aryl hydrocarbon receptor (AhR), a ligand-dependent transcription factor mediates responses to environment stimuli, is emerging as a unique pleiotropic target. Herein, a combined molecular simulation and in vitro investigations identified 3-(3-fluorophenyl)-1H-pyrazolo[3,4-b]pyridine (3FPP) as a novel AhR ligand in T47D and MDA-MB-231 breast cancer cells. Its agonistic effects induced formation of the AhR-AhR nuclear translocator (Arnt) heterodimer and prompted its binding to the penta-nucleotide sequence, called xenobiotic-responsive element (XRE) motif. Moreover, 3FPP augmented the promoter-driven luciferase activities and expression of AhR-regulated genes encoding cytochrome P450 1A1 (CYP1A1) and microRNA (miR)-212/132 cluster. It reduced cell viability, migration, and invasion of both cell lines through AhR signaling. These anticancer properties were concomitant with reduced levels of B-cell lymphoma 2 (BCL-2), SRY-related HMG-box4 (SOX4), snail family zinc finger 2 (SNAI2), and cadherin 2 (CDH2). In vivo, 3FPP suppressed tumor growth and activated AhR signaling in an orthotopic mouse model. In conclusion, our results introduce the fused pyrazolopyridine 3FPP as a novel AhR agonist with AhR-specific anti-breast cancer potential in vitro and in vivo.
Collapse
Affiliation(s)
- Maisa S Abduh
- Immune Responses in Different Diseases Research Group, Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Osama I Alwassil
- Department of Pharmaceutical Sciences, College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11451, Saudi Arabia.
| | - Saleh M Aldaqal
- Immune Responses in Different Diseases Research Group, Department of Surgery, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Manal A Alfwuaires
- Department of Biological Sciences, College of Science, King Faisal University, Hofuf 31982, Saudi Arabia.
| | - Mahdi Farhan
- International Medical Research Center (iMReC), Aqaba 77110, Jordan; Drug Development Department, UniTechPharma, Fribourg 1700, Switzerland.
| | - Hamza Hanieh
- International Medical Research Center (iMReC), Aqaba 77110, Jordan; Basic Medical Sciences Department, Faculty of Medicine, Aqaba Medical Sciences University, Aqaba 77110, Jordan.
| |
Collapse
|
2
|
Liang S, Bo H, Zhang Y, Zhen H, Zhong L. Alizarin, an Agonist of AHR Receptor, Enhances CYP1A1 Enzyme Activity and Induces Transcriptional Changes in Hepatoma Cells. Molecules 2023; 28:7373. [PMID: 37959792 PMCID: PMC10650112 DOI: 10.3390/molecules28217373] [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: 09/24/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
The phytopigment alizarin was previously characterized as an anti-tumor drug owing to its antioxidant or antigenotoxic activities. However, the safety of alizarin is currently still under dispute. In this study, we explored the activity of alizarin in the AHR-CYP1A1 pathway and analyzed the transcriptional changes affected by alizarin using human hepatoma cell line HepG2-based assays. The results showed that alizarin decreased HepG2 cell viability in a dose-dependent manner, with IC50 values between 160.4 and 216.8 μM. Furthermore, alizarin significantly upregulated the expression of CYP1A1 and increased the ethoxyresorufin-O-deethylase activity. Alizarin also exhibited agonistic activity toward the AHR receptor in the XRE-mediated luciferase reporter gene assay, which was further confirmed via the molecular docking assay. In addition, the transcriptional analysis indicated that alizarin may act as a potential carcinogen through significantly enriching several items related to cancer in both DO and KEGG analysis. In brief, our findings indicated that alizarin shows agonistic activities to the AHR receptor through activating the AHR-CYP1A1 signaling pathway in HepG2 cells, which may lead to the risks for cancer developing.
Collapse
Affiliation(s)
- Shengxian Liang
- Institute of Life Sciences and Green Development, College of Life Sciences, Hebei University, Baoding 071000, China; (H.B.); (Y.Z.); (H.Z.)
| | - Haimei Bo
- Institute of Life Sciences and Green Development, College of Life Sciences, Hebei University, Baoding 071000, China; (H.B.); (Y.Z.); (H.Z.)
| | - Yue Zhang
- Institute of Life Sciences and Green Development, College of Life Sciences, Hebei University, Baoding 071000, China; (H.B.); (Y.Z.); (H.Z.)
| | - Hongcheng Zhen
- Institute of Life Sciences and Green Development, College of Life Sciences, Hebei University, Baoding 071000, China; (H.B.); (Y.Z.); (H.Z.)
| | - Li Zhong
- Institute of Life Sciences and Green Development, College of Life Sciences, Hebei University, Baoding 071000, China; (H.B.); (Y.Z.); (H.Z.)
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
3
|
Hanieh H, Bani Ismail M, Alfwuaires MA, Ibrahim HIM, Farhan M. Aryl Hydrocarbon Receptor as an Anticancer Target: An Overview of Ten Years Odyssey. Molecules 2023; 28:molecules28103978. [PMID: 37241719 DOI: 10.3390/molecules28103978] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/22/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
Aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor belonging to the basic helix-loop-helix (bHLH)/per-Arnt-sim (PAS) superfamily, is traditionally known to mediate xenobiotic metabolism. It is activated by structurally diverse agonistic ligands and regulates complicated transcriptional processes through its canonical and non-canonical pathways in normal and malignant cells. Different classes of AhR ligands have been evaluated as anticancer agents in different cancer cells and exhibit efficiency, which has thrust AhR into the limelight as a promising molecular target. There is strong evidence demonstrating the anticancer potential of exogenous AhR agonists including synthetic, pharmaceutical, and natural compounds. In contrast, several reports have indicated inhibition of AhR activity by antagonistic ligands as a potential therapeutic strategy. Interestingly, similar AhR ligands exert variable anticancer or cancer-promoting potential in a cell- and tissue-specific mode of action. Recently, ligand-mediated modulation of AhR signaling pathways and the associated tumor microenvironment is emerging as a potential approach for developing cancer immunotherapeutic drugs. This article reviews advances of AhR in cancer research covering publication from 2012 to early 2023. It summarizes the therapeutic potential of various AhR ligands with an emphasis on exogenous ligands. It also sheds light on recent immunotherapeutic strategies involving AhR.
Collapse
Affiliation(s)
- Hamza Hanieh
- Basic Medical Sciences Department, Faculty of Medicine, Aqaba Medical Sciences University, Aqaba 77110, Jordan
- International Medical Research Center (iMReC), Aqaba 77110, Jordan
| | - Mohammad Bani Ismail
- Basic Medical Sciences Department, Faculty of Medicine, Aqaba Medical Sciences University, Aqaba 77110, Jordan
| | - Manal A Alfwuaires
- Department of Biological Sciences, College of Science, King Faisal University, Hofuf 31982, Saudi Arabia
| | - Hairul-Islam M Ibrahim
- Department of Biological Sciences, College of Science, King Faisal University, Hofuf 31982, Saudi Arabia
| | - Mahdi Farhan
- International Medical Research Center (iMReC), Aqaba 77110, Jordan
- Department of Drug Development, UniTechPharma, 1700 Fribourg, Switzerland
| |
Collapse
|
4
|
Wang LG, Montaño AR, Combs JR, McMahon NP, Solanki A, Gomes MM, Tao K, Bisson WH, Szafran DA, Samkoe KS, Tichauer KM, Gibbs SL. OregonFluor enables quantitative intracellular paired agent imaging to assess drug target availability in live cells and tissues. Nat Chem 2023; 15:729-739. [PMID: 36997700 DOI: 10.1038/s41557-023-01173-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/27/2023] [Indexed: 04/30/2023]
Abstract
Non-destructive fluorophore diffusion across cell membranes to provide an unbiased fluorescence intensity readout is critical for quantitative imaging applications in live cells and tissues. Commercially available small-molecule fluorophores have been engineered for biological compatibility, imparting high water solubility by modifying rhodamine and cyanine dye scaffolds with multiple sulfonate groups. The resulting net negative charge, however, often renders these fluorophores cell-membrane-impermeant. Here we report the design and development of our biologically compatible, water-soluble and cell-membrane-permeable fluorophores, termed OregonFluor (ORFluor). By adapting previously established ratiometric imaging methodology using bio-affinity agents, it is now possible to use small-molecule ORFluor-labelled therapeutic inhibitors to quantitatively visualize their intracellular distribution and protein target-specific binding, providing a chemical toolkit for quantifying drug target availability in live cells and tissues.
Collapse
Affiliation(s)
- Lei G Wang
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR, USA
| | - Antonio R Montaño
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR, USA
| | - Jason R Combs
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR, USA
| | - Nathan P McMahon
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR, USA
| | - Allison Solanki
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR, USA
| | - Michelle M Gomes
- Cancer Early Detection Advanced Research Center (CEDAR), Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Kai Tao
- Cancer Early Detection Advanced Research Center (CEDAR), Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - William H Bisson
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Dani A Szafran
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR, USA
| | - Kimberley S Samkoe
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
- Department of Surgery, Dartmouth Health, Lebanon, NH, USA
| | - Kenneth M Tichauer
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, USA
| | - Summer L Gibbs
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR, USA.
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
5
|
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: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [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
|
6
|
Rosales-Hernández MC, Bello M, Toledano JV, Feregrino BCE, Correa Basurto J, Fragoso Morales LG, Torres-Ramos MA. Molecular dynamics simulations depict structural motions of the whole human aryl hydrocarbon receptor influencing its binding of ligands and HSP90. J Biomol Struct Dyn 2023; 41:13138-13153. [PMID: 36705144 DOI: 10.1080/07391102.2023.2171132] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 01/15/2023] [Indexed: 01/28/2023]
Abstract
The aryl hydrocarbon receptor (AhR) has broad biological functions when its ligands activate it; the non-binding interactions with AhR have not been fully elucidated due to the absence of a complete tridimensional (3D) structure. Therefore, utilization of the whole 3D structure from Homo sapiens AhR by in silico studies will allow us to better study and analyze the binding mode of its full and partial agonists, and antagonists, as well as its interaction with the HSP90 chaperone. The 3D AhR structure was obtained from I-TASSER and subjected to molecular dynamics (MD) simulations to obtain different structural conformations and determine the most populated AhR conformer by clustering analyses. The AhR-3D structures selected from MD simulations and those from clustering analyses were used to achieve docking studies with some of its ligands and protein-protein docking with HSP90. Once the AhR-3D structure was built, its Ramachandran maps and energy showed a well-qualified 3D model. MD simulations showed that the per-Arnt-Sim homology (PAS) PAS A, PAS B, and Q domains underwent conformational changes, identifying the conformation when agonists were binding also, and HSP90 was binding near the PAS A, PAS B, and Q domains. However, when antagonists are binding, HSP90 does not bind near the PAS A, PAS B, and Q domains. These studies show that the complex agonist-AhR-HSP90 can be formed, but this complex is not formed when an antagonist is binding. Knowing the conformations when the ligands bind to AHR and the behavior of HSP90 allows for an understanding of its activity.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Martha Cecilia Rosales-Hernández
- Laboratorio de Biofísica y Biocatálisis, Sección de Estudios de Posgrago e Investigación. Escuela Superior de Medicina, Instituto Politécnico Nacional. Plan de San Luis y Díaz Mirón s/n, Ciudad de México, Mexico
| | - Martiniano Bello
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Seccion de Estudios de Posgrado. Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, México City, Mexico
| | - Jazziel Velazquez Toledano
- Laboratorio de Biofísica y Biocatálisis, Sección de Estudios de Posgrago e Investigación. Escuela Superior de Medicina, Instituto Politécnico Nacional. Plan de San Luis y Díaz Mirón s/n, Ciudad de México, Mexico
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Seccion de Estudios de Posgrado. Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, México City, Mexico
| | - Barbara Citlali Escudero Feregrino
- Laboratorio de Biofísica y Biocatálisis, Sección de Estudios de Posgrago e Investigación. Escuela Superior de Medicina, Instituto Politécnico Nacional. Plan de San Luis y Díaz Mirón s/n, Ciudad de México, Mexico
- Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City, México
| | - José Correa Basurto
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Seccion de Estudios de Posgrado. Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, México City, Mexico
| | - Leticia Guadalupe Fragoso Morales
- Laboratorio de Biofísica y Biocatálisis, Sección de Estudios de Posgrago e Investigación. Escuela Superior de Medicina, Instituto Politécnico Nacional. Plan de San Luis y Díaz Mirón s/n, Ciudad de México, Mexico
| | | |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Fu S, Liao L, Yang Y, Bai Y, Zeng Y, Wang H, Wen J. The pharmacokinetics profiles, pharmacological properties, and toxicological risks of dehydroevodiamine: A review. Front Pharmacol 2022; 13:1040154. [PMID: 36467053 PMCID: PMC9715618 DOI: 10.3389/fphar.2022.1040154] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 11/03/2022] [Indexed: 01/10/2024] Open
Abstract
Dehydroevodiamine (DHE) is a quinazoline alkaloid isolated from Evodiae Fructus (EF, Wuzhuyu in Chinese, Rutaceae family), a well-known traditional Chinese medicine (TCM) which is clinically applied to treat headache, abdominal pain, menstrual pain, abdominal distension, vomiting, acid regurgitation, etc. Modern research demonstrates that DHE is one of the main components of EF. In recent years, DHE has received extensive attention due to its various pharmacological activities. This review is the first to comprehensively summarize the current studies on pharmacokinetics profiles, pharmacological properties, and toxicological risks of DHE in diverse diseases. Pharmacokinetic studies have shown that DHE has a relatively good oral absorption effect in the mean concentration curves in rat plasma and high absorption in the gastrointestinal tract. In addition, distribution re-absorption and enterohepatic circulation may lead to multiple blood concentration peaks of DHE in rat plasma. DHE possesses a wide spectrum of pharmacological properties in the central nervous system, cardiovascular system, and digestive system. Moreover, DHE has anti-inflammatory effects via downregulating pro-inflammatory cytokines and inflammatory mediators. Given the favorable pharmacological activity, DHE is expected to be a potential drug candidate for the treatment of Alzheimer's disease, chronic stress, amnesia, chronic atrophic gastritis, gastric ulcers, and rheumatoid arthritis. In addition, toxicity studies have suggested that DHE has proarrhythmic effects and can impair bile acid homeostasis without causing hepatotoxicity. However, further rigorous and well-designed studies are needed to elucidate the pharmacokinetics, pharmacological effects, potential biological mechanisms, and toxicity of DHE.
Collapse
Affiliation(s)
- Shubin Fu
- Jiujiang Inspection and Testing Certification Center, Jiujiang, China
| | - Liying Liao
- Jiujiang Inspection and Testing Certification Center, Jiujiang, China
| | - Yi Yang
- School of Food and Bioengineering, Xihua University, Chengdu, China
| | - Yan Bai
- School of Food and Bioengineering, Xihua University, Chengdu, China
| | - Yan Zeng
- School of Food and Bioengineering, Xihua University, Chengdu, China
| | - Haoyu Wang
- School of Food and Bioengineering, Xihua University, Chengdu, China
| | - Jianxia Wen
- School of Food and Bioengineering, Xihua University, Chengdu, China
| |
Collapse
|
10
|
Elson DJ, Nguyen BD, Wood R, Zhang Y, Puig-Sanvicens V, Kolluri SK. The cyclin-dependent kinase inhibitor p27 Kip1 interacts with the aryl hydrocarbon receptor and negatively regulates its transcriptional activity. FEBS Lett 2022; 596:2056-2071. [PMID: 35735777 DOI: 10.1002/1873-3468.14434] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 05/09/2022] [Accepted: 05/11/2022] [Indexed: 11/11/2022]
Abstract
p27Kip1 functions to coordinate cell cycle progression through the inhibition of cyclin-dependent kinase (CDK) complexes. p27Kip1 also exerts distinct activities beyond CDK-inhibition, including functioning as a transcriptional regulator. The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor with diverse biological roles. The regulatory inputs that control AhR-mediated transcriptional responses are an active area of investigation. AhR was previously established as a direct regulator of p27Kip1 transcription. Here, we report the physical interaction of AhR and p27Kip1 and show that p27Kip1 expression negatively regulates AhR-mediated transcription. p27Kip1 knockout cells display increased AhR nuclear localisation and significantly higher expression of AhR target genes. This work thus identifies new regulatory cross-talk between p27Kip1 and AhR.
Collapse
Affiliation(s)
- Daniel J Elson
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, USA
| | - Bach D Nguyen
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, USA
| | - Rhand Wood
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, USA
| | - Yi Zhang
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, USA
| | - Veronica Puig-Sanvicens
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, USA
| | - Siva K Kolluri
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, USA.,Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
| |
Collapse
|
11
|
Safe S, Jayaraman A, Chapkin RS. Ah receptor ligands and their impacts on gut resilience: structure-activity effects. Crit Rev Toxicol 2020; 50:463-473. [PMID: 32597352 DOI: 10.1080/10408444.2020.1773759] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD, dioxin) and structurally related halogenated aromatics modulate gene expression and induce biochemical and toxic responses that are mediated by initial binding to the aryl hydrocarbon receptor (AhR). The AhR also binds structurally diverse compound including pharmaceuticals, endogenous biochemicals, health-promoting phytochemicals, and microbial metabolites. Many of these AhR ligands do not induce TCDD-like toxic responses and some AhR ligands such as microbial metabolites of tryptophan play a role in maintaining gut health and protecting against intestinal inflammation and cancer. Many AhR ligands exhibit tissue- and response-specific AhR agonist or antagonist activities, and act as selective AhR modulators (SAhRMs) and this SAhRM-like activity has also been observed in AhR-ligand-mediated effects in the intestine. This review summarizes studies showing that several AhR ligands including phytochemicals and TCDD protect against dextran sodium sulfate-induced intestinal inflammation. In contrast, AhR ligands such as oxazole compounds enhance intestinal inflammation suggesting that AhR-mediated gut health can be enhanced or decreased by selective AhR modulators and this needs to be considered in development of AhR ligands for therapeutic applications in treating intestinal inflammation.
Collapse
Affiliation(s)
- Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, USA
| | - Arul Jayaraman
- Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Robert S Chapkin
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX, USA
| |
Collapse
|
12
|
Artyukov AA, Zelepuga EA, Bogdanovich LN, Lupach NM, Novikov VL, Rutckova TA, Kozlovskaya EP. Marine Polyhydroxynaphthoquinone, Echinochrome A: Prevention of Atherosclerotic Inflammation and Probable Molecular Targets. J Clin Med 2020; 9:E1494. [PMID: 32429179 PMCID: PMC7291202 DOI: 10.3390/jcm9051494] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/06/2020] [Accepted: 05/13/2020] [Indexed: 12/16/2022] Open
Abstract
The effect of low doses of echinochrome A (EchA), a natural polyhydroxy-1,4-naphthoquinone pigment from the sea urchin Scaphechinus mirabilis, has been studied in clinical trials, when it was used as an active substance of the drug Histochrome® and biologically active supplement Thymarin. Several parameters of lipid metabolism, antioxidant status, and the state of the immune system were analyzed in patients with cardiovascular diseases (CVD), including contaminating atherosclerosis. It has been shown that EchA effectively normalizes lipid metabolism, recovers antioxidant status and reduces atherosclerotic inflammation, regardless of the method of these preparations' administrations. Treatment of EchA has led to the stabilization of patients, improved function of the intracellular matrix and decreased epithelial dysfunction. The increased expression of surface human leukocyte antigen DR isotype (HLA-DR) receptors reflects the intensification of intercellular cooperation of immune cells, as well as an increase in the efficiency of processing and presentation of antigens, while the regulation of CD95 + expression levels suggests the stimulation of cell renewal processes. The immune system goes to a different level of functioning. Computer simulations suggest that EchA, with its aromatic structure of the naphthoquinone nucleus, may be a suitable ligand of the cytosolic aryl cell receptor, which affects the response of the immune system and causes the rapid expression of detoxification enzymes such as CYP and DT diaphorase, which play a protective role with CVD. Therefore, EchA possesses not only an antiradical effect and antioxidant activity, but is also a SOD3 mimetic, producing hydrogen peroxide and controlling the expression of cell enzymes through hypoxia-inducible factors (HIF), peroxisome proliferator-activated receptors (PPARs) and aryl hydrocarbon receptor (AhR).
Collapse
Affiliation(s)
- Aleksandr A. Artyukov
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Science, 159 Prospect 100-letiya Vladivostoka, Vladivostok 690022, Russia; (A.A.A.); (E.A.Z.); (V.L.N.); (T.A.R.)
| | - Elena A. Zelepuga
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Science, 159 Prospect 100-letiya Vladivostoka, Vladivostok 690022, Russia; (A.A.A.); (E.A.Z.); (V.L.N.); (T.A.R.)
| | - Larisa N. Bogdanovich
- Medical Association of the Far Eastern Branch of the Russian Academy of Sciences (FEB RAS MO), Kirov Str., 95, Vladivostok 690022, Russia;
| | - Natalia M. Lupach
- Primorye Regional Clinical Hospital No. One (SHI), Aleutskaya Str., 57, Vladivostok, Primorsky Krai 690091, Russia;
| | - Vyacheslav L. Novikov
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Science, 159 Prospect 100-letiya Vladivostoka, Vladivostok 690022, Russia; (A.A.A.); (E.A.Z.); (V.L.N.); (T.A.R.)
| | - Tatyana A. Rutckova
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Science, 159 Prospect 100-letiya Vladivostoka, Vladivostok 690022, Russia; (A.A.A.); (E.A.Z.); (V.L.N.); (T.A.R.)
| | - Emma P. Kozlovskaya
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Science, 159 Prospect 100-letiya Vladivostoka, Vladivostok 690022, Russia; (A.A.A.); (E.A.Z.); (V.L.N.); (T.A.R.)
| |
Collapse
|
13
|
Gourronc FA, Perdew GH, Robertson LW, Klingelhutz AJ. PCB126 blocks the thermogenic beiging response of adipocytes. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:8897-8904. [PMID: 31721030 PMCID: PMC7098842 DOI: 10.1007/s11356-019-06663-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 10/01/2019] [Indexed: 05/16/2023]
Abstract
Subcutaneous white adipose tissue is capable of becoming thermogenic in a process that is referred to as "beiging." Beiging is associated with activation of the uncoupling protein, UCP1, and is known to be important for preventing adipose hypertrophy and development of insulin resistance. Polychlorinated biphenyls (PCBs) accumulate in fat, and it is hypothesized that disruption of adipogenesis and adipocyte function by PCBs may be causative in the development of obesity and diabetes. We developed immortal human subcutaneous preadipocytes that, when differentiated, are capable of beiging. Preadipocytes that were treated with polychlorinated biphenyl congener 126 (PCB126), followed by differentiation, were suppressed for their ability to activate UCP1 upon β-adrenergic stimulation with norepinephrine (NE), demonstrating a block in the beiging response. Treatment of preadipocytes with another known endogenous AhR agonist, indoxyl sulfate (IS), followed by differentiation also blocked the NE-stimulated upregulation of UCP1. Knockdown of the aryl hydrocarbon receptor (AhR) caused the preadipocytes to be refractory to PCB126 and IS effects. The chemical AhR antagonist, CH223191, was effective at preventing the effects of PCB126 but not IS, indicating AhR ligand specificity of CH223191. Repression of NE-induced UCP1 upregulation was also observed when already-differentiated mature adipocytes were treated with PCB126 but not IS. These results indicate that exposure of preadipocytes to endogenous (IS) or exogenous (PCB126) AhR agonists is effective at blocking them from becoming functional adipocytes that are capable of the beiging response. Mature adipocytes may have differential responses. This finding suggests a mechanism by which dioxin-like PCBs such as PCB126 could lead to disruption in energy homeostasis, potentially leading to obesity and diabetes.
Collapse
Affiliation(s)
- Francoise A Gourronc
- Department of Microbiology and Immunology, University of Iowa, 3-612 BSB, 51 Newton Road, Iowa City, IA, 52242, USA
| | - Gary H Perdew
- Center for Molecular Toxicology and Carcinogenesis and the Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, 16802, PA, USA
| | - Larry W Robertson
- Department of Occupational & Environmental Health, College of Public Health, University of Iowa, Iowa City, IA, 52242, USA
| | - Aloysius J Klingelhutz
- Department of Microbiology and Immunology, University of Iowa, 3-612 BSB, 51 Newton Road, Iowa City, IA, 52242, USA.
| |
Collapse
|
14
|
Liu Y, Wei Y, Zhang S, Yan X, Zhu H, Xu L, Zhao B, Xie HQ, Yan B. Regulation of Aryl Hydrocarbon Receptor Signaling Pathway and Dioxin Toxicity by Novel Agonists and Antagonists. Chem Res Toxicol 2020; 33:614-624. [PMID: 31878777 DOI: 10.1021/acs.chemrestox.9b00431] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Dioxins, mostly through activation of aryl hydrocarbon receptor (AhR), are potent toxic substances widely distributed in the environment, while moderated suppression of AhR also exhibits anti-tumor effects. Therefore, the proper modulation of AhR activity may counteract AhR-mediated toxicities and certain diseases. In this investigation, we identified several novel AhR moderate agonists and antagonists using chemical biology approaches. The mechanisms and mode of interactions with AhR by these hits were also revealed using both experimental and computational studies. The newly identified AhR moderate agonists and antagonists were predicted to bind to AhR and modulate AhR signaling. The structure-activity relationships of moderate agonists and antagonists and their unique binding features with AhR have created a solid framework for further optimization of the next generation of AhR modulators.
Collapse
Affiliation(s)
- Yin Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences , Chinese Academy of Sciences , Beijing 100085 , China.,University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Yongyi Wei
- Institute of Environmental Research at Greater Bay, Guangzhou University, Key Laboratory for Water Quality and Conservation of the Pearl River Delta , Ministry of Education , Guangzhou 510006 , China.,School of Environmental Science and Engineering , Shandong University , Qingdao 266237 , China
| | - Songyan Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences , Chinese Academy of Sciences , Beijing 100085 , China.,University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Xiliang Yan
- School of Environmental Science and Engineering , Shandong University , Qingdao 266237 , China.,The Rutgers Center for Computational and Integrative Biology , Camden , New Jersey 08102 , United States
| | - Hao Zhu
- The Rutgers Center for Computational and Integrative Biology , Camden , New Jersey 08102 , United States
| | - Li Xu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences , Chinese Academy of Sciences , Beijing 100085 , China.,University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Bin Zhao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences , Chinese Academy of Sciences , Beijing 100085 , China.,University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Heidi Qunhui Xie
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences , Chinese Academy of Sciences , Beijing 100085 , China.,University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Bing Yan
- Institute of Environmental Research at Greater Bay, Guangzhou University, Key Laboratory for Water Quality and Conservation of the Pearl River Delta , Ministry of Education , Guangzhou 510006 , China.,School of Environmental Science and Engineering , Shandong University , Qingdao 266237 , China
| |
Collapse
|
15
|
Baker JR, Sakoff JA, McCluskey A. The aryl hydrocarbon receptor (AhR) as a breast cancer drug target. Med Res Rev 2019; 40:972-1001. [PMID: 31721255 DOI: 10.1002/med.21645] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/04/2019] [Accepted: 10/29/2019] [Indexed: 12/25/2022]
Abstract
Breast cancer is the most common cancer in women, with more than 1.7 million diagnoses worldwide per annum. Metastatic breast cancer remains incurable, and the presence of triple-negative phenotypes makes targeted treatment impossible. The aryl hydrocarbon receptor (AhR), most commonly associated with the metabolism of xenobiotic ligands, has emerged as a promising biological target for the treatment of this deadly disease. Ligands for the AhR can be classed as exogenous or endogenous and may have agonistic or antagonistic activity. It has been well reported that agonistic ligands may have potent and selective growth inhibition activity in a number of oncogenic cell lines, and one (aminoflavone) has progressed to phase I clinical trials for breast cancer sufferers. In this study, we examine the current state of the literature in this area and elucidate the promising advances that are being made in hijacking the cytosolic-to-nuclear pathway of the AhR for the possible future treatment of breast cancer.
Collapse
Affiliation(s)
- Jennifer R Baker
- Chemistry, School of Environmental & Life Sciences, the University of Newcastle, Callaghan, NSW, Australia
| | - Jennette A Sakoff
- Department of Medical Oncology, Calvary Mater Newcastle Hospital, Waratah, NSW, Australia
| | - Adam McCluskey
- Chemistry, School of Environmental & Life Sciences, the University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
16
|
Biagioli M, Carino A, Fiorucci C, Annunziato G, Marchianò S, Bordoni M, Roselli R, Giorgio CD, Castiglione F, Ricci P, Bruno A, Faccini A, Distrutti E, Baldoni M, Costantino G, Fiorucci S. The Aryl Hydrocarbon Receptor (AhR) Mediates the Counter-Regulatory Effects of Pelargonidins in Models of Inflammation and Metabolic Dysfunctions. Nutrients 2019; 11:E1820. [PMID: 31394746 PMCID: PMC6723439 DOI: 10.3390/nu11081820] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/02/2019] [Accepted: 08/05/2019] [Indexed: 12/15/2022] Open
Abstract
Pelargonidins are anthocyanidins thought to be beneficial for the human health, although controversies exist over the doses needed and the unclear mechanism of action, along with poor systemic bioavailability. One putative target of pelargonidins is the aryl hydrocarbon receptor (AhR). A synthetic pelargonidin (Mt-P) was synthesized by the methylation of the pelargonidin (the natural compound indicated as P). Mt-P transactivated the AhR with an EC50 of 1.97 µM and was ~2-fold more potent than the natural compound. In vitro Mt-P attenuated pro-inflammatory activities of Raw264.7 macrophage cells in an AhR-dependent manner. In vivo, administration of the Mt-P in Balb/c mice resulted in a dose-dependent attenuation of signs and symptoms of colitis induced by TNBS. A dose of 5 mg/kg Mt-P, but not the natural compound P, reversed intestinal inflammation and increased expression of Tnf-α, Ifn-ƴ, and Il-6, while promoted the expansion of regulatory T cells and M2 macrophages. In C57BL/6J mice fed a high fat diet (HFD), Mt-P attenuated body weight gain, intestinal and liver inflammation, and ameliorated insulin sensitivity, while worsened liver steatosis by up-regulating the liver expression of Cd36 and Apo100b. These effects were abrogated by AhR gene ablation. Mt-P is a synthetic pelargonidin endowed with robust AhR agonist activity that exerts beneficial effects in murine models of inflammation and metabolic dysfunction.
Collapse
Affiliation(s)
- Michele Biagioli
- Dipartimento di Scienze Biomediche e Chirurgiche, Università di Perugia, 06100 Perugia, Italy
| | - Adriana Carino
- Dipartimento di Scienze Biomediche e Chirurgiche, Università di Perugia, 06100 Perugia, Italy
| | - Chiara Fiorucci
- Dipartimento di Scienze Biomediche e Chirurgiche, Università di Perugia, 06100 Perugia, Italy
| | | | - Silvia Marchianò
- Dipartimento di Scienze Biomediche e Chirurgiche, Università di Perugia, 06100 Perugia, Italy
| | - Martina Bordoni
- Dipartimento di Scienze Biomediche e Chirurgiche, Università di Perugia, 06100 Perugia, Italy
| | - Rosalinda Roselli
- Department of Pharmacy, University of Naples Federico II, 80100 Naples, Italy
| | - Cristina Di Giorgio
- Dipartimento di Scienze Biomediche e Chirurgiche, Università di Perugia, 06100 Perugia, Italy
| | - Federica Castiglione
- Dipartimento di Scienze Biomediche e Chirurgiche, Università di Perugia, 06100 Perugia, Italy
| | - Patrizia Ricci
- Dipartimento di Scienze Biomediche e Chirurgiche, Università di Perugia, 06100 Perugia, Italy
| | - Agostino Bruno
- Department of Food and Drugs, University of Parma, 43100 Parma, Italy
| | - Andrea Faccini
- Centro Interdipartimentale Misure 'G. Casnati'-University of Parma, 43100 Parma, Italy
| | | | - Monia Baldoni
- Dipartimento di Scienze Biomediche e Chirurgiche, Università di Perugia, 06100 Perugia, Italy
| | | | - Stefano Fiorucci
- Dipartimento di Scienze Biomediche e Chirurgiche, Università di Perugia, 06100 Perugia, Italy.
| |
Collapse
|
17
|
Balaguer P, Delfosse V, Bourguet W. Mechanisms of endocrine disruption through nuclear receptors and related pathways. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.coemr.2019.04.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
18
|
The Henna pigment Lawsone activates the Aryl Hydrocarbon Receptor and impacts skin homeostasis. Sci Rep 2019; 9:10878. [PMID: 31350436 PMCID: PMC6659674 DOI: 10.1038/s41598-019-47350-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 07/15/2019] [Indexed: 12/20/2022] Open
Abstract
As a first host barrier, the skin is constantly exposed to environmental insults that perturb its integrity. Tight regulation of skin homeostasis is largely controlled by the aryl hydrocarbon receptor (AhR). Here, we demonstrate that Henna and its major pigment, the naphthoquinone Lawsone activate AhR, both in vitro and in vivo. In human keratinocytes and epidermis equivalents, Lawsone exposure enhances the production of late epidermal proteins, impacts keratinocyte differentiation and proliferation, and regulates skin inflammation. To determine the potential use of Lawsone for therapeutic application, we harnessed human, murine and zebrafish models. In skin regeneration models, Lawsone interferes with physiological tissue regeneration and inhibits wound healing. Conversely, in a human acute dermatitis model, topical application of a Lawsone-containing cream ameliorates skin irritation. Altogether, our study reveals how a widely used natural plant pigment is sensed by the host receptor AhR, and how the physiopathological context determines beneficial and detrimental outcomes.
Collapse
|
19
|
Giani Tagliabue S, Faber SC, Motta S, Denison MS, Bonati L. Modeling the binding of diverse ligands within the Ah receptor ligand binding domain. Sci Rep 2019; 9:10693. [PMID: 31337850 PMCID: PMC6650409 DOI: 10.1038/s41598-019-47138-z] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 07/02/2019] [Indexed: 01/08/2023] Open
Abstract
The Ah receptor (AhR) is a ligand-dependent transcription factor belonging to the basic helix-loop-helix Per-Arnt-Sim (bHLH-PAS) superfamily. Binding to and activation of the AhR by a variety of chemicals results in the induction of expression of diverse genes and production of a broad spectrum of biological and toxic effects. The AhR also plays important roles in several physiological responses, which has led it to become a novel target for the development of therapeutic drugs. Differences in the interactions of various ligands within the AhR ligand binding domain (LBD) may contribute to differential modulation of AhR functionality. We combined computational and experimental analyses to investigate the binding modes of a group of chemicals representative of major classes of AhR ligands. On the basis of a novel computational approach for molecular docking to the homology model of the AhR LBD that includes the receptor flexibility, we predicted specific residues within the AhR binding cavity that play a critical role in binding of three distinct groups of chemicals. The prediction was validated by site-directed mutagenesis and evaluation of the relative ligand binding affinities for the mutant AhRs. These results provide an avenue for understanding ligand modulation of the AhR functionality and for rational drug design.
Collapse
Affiliation(s)
- Sara Giani Tagliabue
- Department of Earth and Environmental Sciences, University of Milano-Bicocca, Milan, Italy
| | - Samantha C Faber
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Stefano Motta
- Department of Earth and Environmental Sciences, University of Milano-Bicocca, Milan, Italy
| | - Michael S Denison
- Department of Environmental Toxicology, University of California, Davis, CA, USA
| | - Laura Bonati
- Department of Earth and Environmental Sciences, University of Milano-Bicocca, Milan, Italy.
| |
Collapse
|
20
|
Identification of the novel role of butyrate as AhR ligand in human intestinal epithelial cells. Sci Rep 2019; 9:643. [PMID: 30679727 PMCID: PMC6345974 DOI: 10.1038/s41598-018-37019-2] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 11/28/2018] [Indexed: 12/18/2022] Open
Abstract
The ligand activated transcription factor, aryl hydrocarbon receptor (AhR) emerged as a critical regulator of immune and metabolic processes in the gastrointestinal tract. In the gut, a main source of AhR ligands derives from commensal bacteria. However, many of the reported microbiota-derived ligands have been restricted to indolyl metabolites. Here, by screening commensal bacteria supernatants on an AhR reporter system expressed in human intestinal epithelial cell line (IEC), we found that the short chain fatty acid (SCFA) butyrate induced AhR activity and the transcription of AhR-dependent genes in IECs. We showed that AhR ligand antagonists reduced the effects of butyrate on IEC suggesting that butyrate could act as a ligand of AhR, which was supported by the nuclear translocation of AhR induced by butyrate and in silico structural modelling. In conclusion, our findings suggest that (i) butyrate activates AhR pathway and AhR-dependent genes in human intestinal epithelial cell-lines (ii) butyrate is a potential ligand for AhR which is an original mechanism of gene regulation by SCFA.
Collapse
|
21
|
Evidence of selective activation of aryl hydrocarbon receptor nongenomic calcium signaling by pyrene. Biochem Pharmacol 2018; 158:1-12. [PMID: 30248327 DOI: 10.1016/j.bcp.2018.09.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 09/20/2018] [Indexed: 12/02/2022]
Abstract
In its classical genomic mode of action, the aryl hydrocarbon receptor (AhR) acts as a ligand activated transcription factor regulating expression of target genes such as CYP1A1 and CYP1B1. Some ligands may also trigger more rapid nongenomic responses through AhR, including calcium signaling (Ca2+). In the present study we observed that pyrene induced a relatively rapid increase in intracellular Ca2+-concentrations ([Ca2+]i) in human microvascular endothelial cells (HMEC-1) and human embryonic kidney cells (HEK293) that was attenuated by AhR-inhibitor treatment and/or transient AhR knockdown by RNAi. In silico molecular docking based on homology models, suggested that pyrene is not able to bind to the human AhR in the agonist conformation. Instead, pyrene docked in the antagonist conformation of the AhR PAS-B binding pocket, although the interaction differed from antagonists such as GNF-351 and CH223191. Accordingly, pyrene did not induce CYP1A1 or CYP1B1, but suppressed CYP1-expression by benzo[a]pyrene (B[a]P) in HMEC-1 cells, confirming that pyrene act as an antagonist of AhR-induced gene expression. Use of pharmacological inhibitors and Ca2+-free medium indicated that the pyrene-induced AhR nongenomic [Ca2+]i increase was initiated by Ca2+-release from intracellular stores followed by a later phase of extracellular Ca2+-influx, consistent with store operated calcium entry (SOCE). These effects was accompanied by an AhR-dependent reduction in ordered membrane lipid domains, as determined by di-4-ANEPPDHQ staining. Addition of cholesterol inhibited both the pyrene-induced [Ca2+]i-increase and alterations in membrane lipid order. In conclusion, we propose that pyrene binds to AhR, act as an antagonist of the canonical genomic AhR/Arnt/CYP1-pathway, reduces ordered membrane lipid domains, and activates AhR nongenomic Ca2+-signaling from intracellular stores.
Collapse
|
22
|
Chlebowski AC, Garcia GR, La Du JK, Bisson WH, Truong L, Massey Simonich SL, Tanguay RL. Mechanistic Investigations Into the Developmental Toxicity of Nitrated and Heterocyclic PAHs. Toxicol Sci 2018; 157:246-259. [PMID: 28186253 PMCID: PMC5414855 DOI: 10.1093/toxsci/kfx035] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Nitrated polycyclic aromatic hydrocarbons (NPAHs) and heterocyclic PAHs (HPAHs) are recognized environmental pollutants. However, the health risks of NPAHs and HPAHs to humans and environmental systems are not well-studied. The developmental zebrafish (Danio rerio) model was used to evaluate the toxicity of a structurally diverse set of 27 NPAHs and 10 HPAHs. The individual activity of each compound towards the aryl hydrocarbon receptor (AHR), including the role of the AHR in observed toxicity, and genetic markers of oxidative stress and cardiac toxicity were evaluated. Zebrafish embryos were exposed from 6 to 120 hours post fertilization (hpf), to a broad concentration range of individual compounds, and evaluated for 22 developmental endpoints. The potential role of AHR was determined using the transgenic Tg(cyp1a:nls-egfp) reporter zebrafish line. All compounds were screened computationally through molecular docking using a previously developed AHR models of zebrafish isoforms 1A, 1B, and 2. Some compounds did not induce observable developmental toxic responses, whereas others produced statistically significant concentration-dependent toxicity. The tested compounds also exhibited a range of predicted AHR binding and cyp1a/GFP induction patterns, including cyp1a expression in the liver, vasculature, skin, and yolk, which we determined to be due to distinct isoforms of the AHR, using morpholino oligonucleotide knockdown. Furthermore, we investigated mRNA expression of oxidative and cardiac stress genes at 48 and 120 hpf, which indicated several potential mechanisms-of-action for NPAHs. Overall, we observed a range of developmental toxicities, cyp1a/GFP expression patterns, and gene expression profiles, suggestive of several potential mechanisms of action.
Collapse
Affiliation(s)
- Anna C Chlebowski
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon, USA
| | - Gloria R Garcia
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon, USA
| | - Jane K La Du
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon, USA
| | - William H Bisson
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon, USA
| | - Lisa Truong
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon, USA
| | - Staci L Massey Simonich
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon, USA.,Department of Chemistry, Oregon State University, Corvallis, Oregon, USA
| | - Robert L Tanguay
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon, USA
| |
Collapse
|
23
|
Zhang Y, Yan T, Sun D, Xie C, Zheng Y, Zhang L, Yagai T, Krausz KW, Bisson WH, Yang X, Gonzalez FJ. Structure-Activity Relationships of the Main Bioactive Constituents of Euodia rutaecarpa on Aryl Hydrocarbon Receptor Activation and Associated Bile Acid Homeostasis. Drug Metab Dispos 2018; 46:1030-1040. [PMID: 29691238 DOI: 10.1124/dmd.117.080176] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 04/19/2018] [Indexed: 12/02/2022] Open
Abstract
Rutaecarpine (RUT), evodiamine (EOD), and dehydroevodiamine (DHED) are the three main bioactive indoloquinazoline alkaloids isolated from Euodia rutaecarpa, a widely prescribed traditional Chinese medicine. Here, the structure-activity relationships of these analogs for aryl hydrocarbon receptor (AHR) activation were explored by use of Ahr-deficient (Ahr-/-) mice, primary hepatocyte cultures, luciferase reporter gene assays, in silico ligand-docking studies, and metabolomics. In vitro, both mRNA analysis of AHR target genes in mouse primary hepatocytes and luciferase reporter assays in hepatocarcinoma cell lines demonstrated that RUT, EOD, and DHED significantly activated AHR, with an efficacy order of RUT > DHED > EOD. Ligand-docking analysis predicted that the methyl substitute at the N-14 atom was a key factor affecting AHR activation. In vivo, EOD was poorly orally absorbed and failed to activate AHR, whereas RUT and DHED markedly upregulated expression of the hepatic AHR gene battery in wild-type mice, but not in Ahr-/- mice. Furthermore, RUT, EOD, and DHED were not hepatotoxic at the doses used; however, RUT and DHED disrupted bile acid homeostasis in an AHR-dependent manner. These findings revealed that the methyl group at the N-14 atom of these analogs and their pharmacokinetic behaviors were the main determinants for AHR activation, and suggest that attention should be given to monitoring bile acid metabolism in the clinical use of E. rutaecarpa.
Collapse
Affiliation(s)
- Youbo Zhang
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (Yo.Z., Ti.Y., D.S. C.X., To.Y., K.W.K., F.J.G.); State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University, Beijing, China (Yo.Z., Yi.Z., L.Z., X.Y.); Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon (W.H.B.); and College of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, Liaoning, China (D.S.)
| | - Tingting Yan
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (Yo.Z., Ti.Y., D.S. C.X., To.Y., K.W.K., F.J.G.); State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University, Beijing, China (Yo.Z., Yi.Z., L.Z., X.Y.); Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon (W.H.B.); and College of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, Liaoning, China (D.S.)
| | - Dongxue Sun
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (Yo.Z., Ti.Y., D.S. C.X., To.Y., K.W.K., F.J.G.); State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University, Beijing, China (Yo.Z., Yi.Z., L.Z., X.Y.); Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon (W.H.B.); and College of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, Liaoning, China (D.S.)
| | - Cen Xie
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (Yo.Z., Ti.Y., D.S. C.X., To.Y., K.W.K., F.J.G.); State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University, Beijing, China (Yo.Z., Yi.Z., L.Z., X.Y.); Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon (W.H.B.); and College of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, Liaoning, China (D.S.)
| | - Yiran Zheng
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (Yo.Z., Ti.Y., D.S. C.X., To.Y., K.W.K., F.J.G.); State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University, Beijing, China (Yo.Z., Yi.Z., L.Z., X.Y.); Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon (W.H.B.); and College of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, Liaoning, China (D.S.)
| | - Lei Zhang
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (Yo.Z., Ti.Y., D.S. C.X., To.Y., K.W.K., F.J.G.); State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University, Beijing, China (Yo.Z., Yi.Z., L.Z., X.Y.); Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon (W.H.B.); and College of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, Liaoning, China (D.S.)
| | - Tomoki Yagai
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (Yo.Z., Ti.Y., D.S. C.X., To.Y., K.W.K., F.J.G.); State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University, Beijing, China (Yo.Z., Yi.Z., L.Z., X.Y.); Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon (W.H.B.); and College of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, Liaoning, China (D.S.)
| | - Kristopher W Krausz
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (Yo.Z., Ti.Y., D.S. C.X., To.Y., K.W.K., F.J.G.); State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University, Beijing, China (Yo.Z., Yi.Z., L.Z., X.Y.); Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon (W.H.B.); and College of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, Liaoning, China (D.S.)
| | - William H Bisson
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (Yo.Z., Ti.Y., D.S. C.X., To.Y., K.W.K., F.J.G.); State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University, Beijing, China (Yo.Z., Yi.Z., L.Z., X.Y.); Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon (W.H.B.); and College of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, Liaoning, China (D.S.)
| | - Xiuwei Yang
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (Yo.Z., Ti.Y., D.S. C.X., To.Y., K.W.K., F.J.G.); State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University, Beijing, China (Yo.Z., Yi.Z., L.Z., X.Y.); Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon (W.H.B.); and College of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, Liaoning, China (D.S.)
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (Yo.Z., Ti.Y., D.S. C.X., To.Y., K.W.K., F.J.G.); State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University, Beijing, China (Yo.Z., Yi.Z., L.Z., X.Y.); Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon (W.H.B.); and College of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, Liaoning, China (D.S.)
| |
Collapse
|
24
|
Ehrlich AK, Pennington JM, Bisson WH, Kolluri SK, Kerkvliet NI. TCDD, FICZ, and Other High Affinity AhR Ligands Dose-Dependently Determine the Fate of CD4+ T Cell Differentiation. Toxicol Sci 2018; 161:310-320. [PMID: 29040756 PMCID: PMC5837604 DOI: 10.1093/toxsci/kfx215] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
FICZ and TCDD, two high-affinity AhR ligands, are reported to have opposite effects on T cell differentiation with TCDD inducing regulatory T cells and FICZ inducing Th17 cells. This dichotomy has been attributed to ligand-intrinsic differences in AhR activation, although differences in sensitivity to metabolism complicate the issue. TCDD is resistant to AhR-induced metabolism and produces sustained AhR activation following a single dose in the μg/kg range, whereas FICZ is rapidly metabolized and AhR activation is transient. Nonetheless, prior studies comparing FICZ with TCDD have generally used the same 10-50 μg/kg dose range, and thus the two ligands would not equivalently activate AhR. We hypothesized that high-affinity AhR ligands can promote CD4+ T cell differentiation into both Th17 cells and Tregs, with fate depending on the extent and duration of AhR activation. We compared the immunosuppressive effects of TCDD and FICZ, along with two other rapidly metabolized ligands (ITE and 11-Cl-BBQ) in an acute alloresponse mouse model. The dose and timing of administration of each ligand was optimized for TCDD-equivalent Cyp1a1 induction. When optimized, all of the ligands suppressed the alloresponse in conjunction with the induction of Foxp3- Tr1 cells on day 2 and the expansion of natural Foxp3+ Tregs on day 10. In contrast, a low dose of FICZ induced transient expression of Cyp1a1 and did not induce Tregs or suppress the alloresponse but enhanced IL-17 production. Interestingly, low doses of the other ligands, including TCDD, also increased IL-17 production on day 10. These findings support the conclusion that the dose and the duration of AhR activation by high-affinity AhR ligands are the primary factors driving the fate of T cell differentiation.
Collapse
Affiliation(s)
- Allison K Ehrlich
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon 97331
| | - Jamie M Pennington
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon 97331
| | - William H Bisson
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon 97331
| | - Siva K Kolluri
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon 97331
| | - Nancy I Kerkvliet
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon 97331
| |
Collapse
|
25
|
Dolciami D, Gargaro M, Cerra B, Scalisi G, Bagnoli L, Servillo G, Fazia MAD, Puccetti P, Quintana FJ, Fallarino F, Macchiarulo A. Binding Mode and Structure-Activity Relationships of ITE as an Aryl Hydrocarbon Receptor (AhR) Agonist. ChemMedChem 2018; 13:270-279. [PMID: 29266750 DOI: 10.1002/cmdc.201700669] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 12/15/2017] [Indexed: 12/13/2022]
Abstract
Discovered as a modulator of the toxic response to environmental pollutants, aryl hydrocarbon receptor (AhR) has recently gained attention for its involvement in various physiological and pathological pathways. AhR is a ligand-dependent transcription factor activated by a large array of chemical compounds, which include metabolites of l-tryptophan (l-Trp) catabolism as endogenous ligands of the receptor. Among these, 2-(1'H-indole-3'-carbonyl)thiazole-4-carboxylic acid methyl ester (ITE) has attracted interest in the scientific community, being endowed with nontoxic, immunomodulatory, and anticancer AhR-mediated functions. So far, no information about the binding mode and interactions of ITE with AhR is available. In this study, we used docking and molecular dynamics to propose a putative binding mode of ITE into the ligand binding pocket of AhR. Mutagenesis studies were then instrumental in validating the proposed binding mode, identifying His 285 and Tyr 316 as important key residues for ligand-dependent receptor activation. Finally, a set of ITE analogues was synthesized and tested to further probe molecular interactions of ITE to AhR and characterize the relevance of specific functional groups in the chemical structure for receptor activity.
Collapse
Affiliation(s)
- Daniela Dolciami
- Department of Pharmaceutical Sciences, University of Perugia, via del Liceo 1, 06123, Perugia, Italy
| | - Marco Gargaro
- Department of Experimental Medicine, University of Perugia, via Gambuli 1, 06132, Perugia, Italy
| | - Bruno Cerra
- Department of Pharmaceutical Sciences, University of Perugia, via del Liceo 1, 06123, Perugia, Italy
| | - Giulia Scalisi
- Department of Experimental Medicine, University of Perugia, via Gambuli 1, 06132, Perugia, Italy
| | - Luana Bagnoli
- Department of Pharmaceutical Sciences, University of Perugia, via del Liceo 1, 06123, Perugia, Italy
| | - Giuseppe Servillo
- Department of Experimental Medicine, University of Perugia, via Gambuli 1, 06132, Perugia, Italy
| | - Maria Agnese Della Fazia
- Department of Experimental Medicine, University of Perugia, via Gambuli 1, 06132, Perugia, Italy
| | - Paolo Puccetti
- Department of Experimental Medicine, University of Perugia, via Gambuli 1, 06132, Perugia, Italy
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Francesca Fallarino
- Department of Experimental Medicine, University of Perugia, via Gambuli 1, 06132, Perugia, Italy
| | - Antonio Macchiarulo
- Department of Pharmaceutical Sciences, University of Perugia, via del Liceo 1, 06123, Perugia, Italy
| |
Collapse
|
26
|
Tkachenko A, Bermudez M, Irmer-Stooff S, Genkinger D, Henkler-Stephani F, Wolber G, Luch A. Nuclear transport of the human aryl hydrocarbon receptor and subsequent gene induction relies on its residue histidine 291. Arch Toxicol 2017; 92:1151-1160. [PMID: 29164305 DOI: 10.1007/s00204-017-2129-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 11/15/2017] [Indexed: 12/11/2022]
Abstract
The aryl hydrocarbon receptor (AHR) is a ligand-dependent transcription factor involved in the metabolism of physiological substances and xenobiotics, representing an interesting target in both toxicology and pharmacology. In this study, we investigated the ligand-dependent conjunction of nuclear import of the human AHR in living cells and target gene induction. Our findings strengthen the theory that the AHR triggers a precisely defined and rapid reaction upon binding to endogenous ligands, while the xenobiotic β-naphthoflavone only induces rather unspecific and slow effects. To better illuminate the ligand-mediated responses of the human AHR, we applied site-directed mutagenesis and identified histidine 291 as key residue for AHR functionality, essential for both nuclear import and target gene induction. Contrary, replacing histidine at position 291 by alanine did not affect nucleo-cytoplasmic shuttling, showing that permanent endogenous import and ligand-induced import of the AHR into the nucleus are two independent and differently regulated processes. Combining these observations with our structural investigations using a homology model of the AHR-PAS B domain, we suggest a dual role of histidine 291: (1) a major role for shaping the ligand binding site including direct interactions with ligands and, (2) an essential role for the conformational dynamics of a PAS B loop, which most likely influences the association of the AHR with the AHR nuclear translocator through interference with their protein-protein interface.
Collapse
Affiliation(s)
- A Tkachenko
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Strasse 8-10, 10589, Berlin, Germany.
| | - M Bermudez
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Straße 2 + 4, 14195, Berlin, Germany
| | - S Irmer-Stooff
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Strasse 8-10, 10589, Berlin, Germany
| | - D Genkinger
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Strasse 8-10, 10589, Berlin, Germany
| | - F Henkler-Stephani
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Strasse 8-10, 10589, Berlin, Germany
| | - G Wolber
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Straße 2 + 4, 14195, Berlin, Germany
| | - A Luch
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Strasse 8-10, 10589, Berlin, Germany
| |
Collapse
|
27
|
Role of the aryl hydrocarbon receptor in carcinogenesis and potential as an anti-cancer drug target. Arch Toxicol 2017; 91:2497-2513. [PMID: 28508231 DOI: 10.1007/s00204-017-1981-2] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 05/08/2017] [Indexed: 12/31/2022]
Abstract
The aryl hydrocarbon receptor (AhR) was initially identified as the receptor that binds and mediates the toxic effects induced by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and structurally related halogenated aromatics. Other toxic compounds including some polynuclear aromatic hydrocarbons act through the AhR; however, during the last 25 years, it has become apparent that the AhR plays an essential role in maintaining cellular homeostasis. Moreover, the scope of ligands that bind the AhR includes endogenous compounds such as multiple tryptophan metabolites, other endogenous biochemicals, pharmaceuticals and health-promoting phytochemicals including flavonoids, indole-3-carbinol and its metabolites. It has also been shown that like other receptors, the AhR is a drug target for multiple diseases including cancer, where both AhR agonists and antagonists effectively block many of the critical hallmarks of cancer in multiple tumor types. This review describes the anti-cancer activities of AhR ligands and demonstrates that it is time to separate the AhR from TCDD and exploit the potential of the AhR as a novel target for cancer chemotherapy.
Collapse
|
28
|
Polyphenolics from mango (Mangifera indica L.) suppress breast cancer ductal carcinoma in situ proliferation through activation of AMPK pathway and suppression of mTOR in athymic nude mice. J Nutr Biochem 2017; 41:12-19. [DOI: 10.1016/j.jnutbio.2016.11.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 10/31/2016] [Accepted: 11/14/2016] [Indexed: 12/17/2022]
|
29
|
Molecular modeling of the AhR structure and interactions can shed light on ligand-dependent activation and transformation mechanisms. CURRENT OPINION IN TOXICOLOGY 2017; 2:42-49. [PMID: 28497129 DOI: 10.1016/j.cotox.2017.01.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Molecular modeling has given important contributions to elucidation of the main stages in the AhR signal transduction pathway. Despite the lack of experimentally determined structures of the AhR functional domains, information derived from homologous systems has been exploited for modeling their structure and interactions. Homology models of the AhR PASB domain have provided information on the binding cavity and contributed to elucidate species-specific differences in ligand binding. Molecular Docking simulations of the ligand binding process have given insights into differences in binding of diverse agonists, antagonists, and selective AhR modulators, and their application to virtual screening of large databases of compounds have allowed identification of novel AhR ligands. Recently available structural information on protein-protein and protein-DNA complexes of other bHLH-PAS systems has opened the way for modeling the AhR:ARNT dimer structure and investigating the mechanisms of AhR transformation and DNA binding. Future research directions should include simulation of the protein dynamics to obtain a more reliable description of intermolecular interactions involved in signal transmission.
Collapse
|
30
|
Miller MF, Goodson WH, Manjili MH, Kleinstreuer N, Bisson WH, Lowe L. Low-Dose Mixture Hypothesis of Carcinogenesis Workshop: Scientific Underpinnings and Research Recommendations. ENVIRONMENTAL HEALTH PERSPECTIVES 2017; 125:163-169. [PMID: 27517672 PMCID: PMC5289915 DOI: 10.1289/ehp411] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 06/21/2016] [Accepted: 07/14/2016] [Indexed: 05/10/2023]
Abstract
BACKGROUND The current single-chemical-as-carcinogen risk assessment paradigm might underestimate or miss the cumulative effects of exposure to chemical mixtures, as highlighted in recent work from the Halifax Project. This is particularly important for chemical exposures in the low-dose range that may be affecting crucial cancer hallmark mechanisms that serve to enable carcinogenesis. OBJECTIVE Could ongoing low-dose exposures to a mixture of commonly encountered environmental chemicals produce effects in concert that lead to carcinogenesis? A workshop held at the NIEHS in August 2015 evaluated the scientific support for the low-dose mixture hypothesis of carcinogenesis and developed a research agenda. Here we describe the science that supports this novel theory, identify knowledge gaps, recommend future methodologies, and explore preventative risk assessment and policy decision-making that incorporates cancer biology, environmental health science, translational toxicology, and clinical epidemiology. DISCUSSION AND CONCLUSIONS The theoretical merits of the low-dose carcinogenesis hypothesis are well founded with clear biological relevance, and therefore, the premise warrants further investigation. Expert recommendations include the need for better insights into the ways in which noncarcinogenic constituents might combine to uniquely affect the process of cellular transformation (in vitro) and environmental carcinogenesis (in vivo), including investigations of the role of key defense mechanisms in maintaining transformed cells in a dormant state. The scientific community will need to acknowledge limitations of animal-based models in predicting human responses; evaluate biological events leading to carcinogenesis both spatially and temporally; examine the overlap between measurable cancer hallmarks and characteristics of carcinogens; incorporate epigenetic biomarkers, in silico modelling, high-performance computing and high-resolution imaging, microbiome, metabolomics, and transcriptomics into future research efforts; and build molecular annotations of network perturbations. The restructuring of many existing regulatory frameworks will require adequate testing of relevant environmental mixtures to build a critical mass of evidence on which to base policy decisions. Citation: Miller MF, Goodson WH III, Manjili MH, Kleinstreuer N, Bisson WH, Lowe L. 2017. Low-Dose Mixture Hypothesis of Carcinogenesis Workshop: scientific underpinnings and research recommendations. Environ Health Perspect 125:163-169; http://dx.doi.org/10.1289/EHP411.
Collapse
Affiliation(s)
- Mark F. Miller
- National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina, USA
- Address correspondence to M.F. Miller, National Institute of Environmental Health Sciences, 111 T.W. Alexander Dr., Research Triangle Park, NC 27709 USA. Telephone: (919) 541-7758. E-mail: , or W.H. Bisson, Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331 USA. Telephone: (541) 737-5735. E-mail:
| | - William H. Goodson
- California Pacific Medical Center Research Institute, San Francisco, California, USA
| | - Masoud H. Manjili
- Department of Microbiology and Immunology, Virginia Commonwealth University, Massey Cancer Center, Richmond, Virginia, USA
| | - Nicole Kleinstreuer
- National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina, USA
| | - William H. Bisson
- Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon, USA
- Address correspondence to M.F. Miller, National Institute of Environmental Health Sciences, 111 T.W. Alexander Dr., Research Triangle Park, NC 27709 USA. Telephone: (919) 541-7758. E-mail: , or W.H. Bisson, Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331 USA. Telephone: (541) 737-5735. E-mail:
| | - Leroy Lowe
- Getting to Know Cancer, Truro, Nova Scotia, Canada
- Lancaster Environment Centre, Lancaster University, Bailrigg, Lancaster, United Kingdom
| |
Collapse
|
31
|
Xue Z, Li D, Yu W, Zhang Q, Hou X, He Y, Kou X. Mechanisms and therapeutic prospects of polyphenols as modulators of the aryl hydrocarbon receptor. Food Funct 2017; 8:1414-1437. [DOI: 10.1039/c6fo01810f] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Polyphenolic AhR modulators displayed concentration-, XRE-, gene-, species- and cell-specific agonistic/antagonistic activity.
Collapse
Affiliation(s)
- Zhaohui Xue
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300072
- China
| | - Dan Li
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300072
- China
| | - Wancong Yu
- Medical Plant Laboratory
- Tianjin Research Center of Agricultural Biotechnology
- Tianjin 3000381
- China
| | - Qian Zhang
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300072
- China
| | - Xiaonan Hou
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300072
- China
| | - Yulong He
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300072
- China
| | - Xiaohong Kou
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300072
- China
| |
Collapse
|
32
|
Conformational modulation of the farnesoid X receptor by prenylflavonoids: Insights from hydrogen deuterium exchange mass spectrometry (HDX-MS), fluorescence titration and molecular docking studies. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2016; 1864:1667-1677. [PMID: 27596062 DOI: 10.1016/j.bbapap.2016.08.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 08/29/2016] [Accepted: 08/31/2016] [Indexed: 12/20/2022]
Abstract
We report on the molecular interactions of the farnesoid X receptor (FXR) with prenylflavonoids, an emerging class of FXR modulators. FXR is an attractive therapeutic target for mitigating metabolic syndromes (MetS) because FXR activates the inhibitory nuclear receptor, small heterodimer partner (SHP), thereby inhibiting both gluconeogenesis and de novo lipogenesis. We and others have shown that xanthohumol (XN), the principal prenylflavonoid of the hop plant (Humulus lupulus L.), is a FXR agonist based on its ability to affect lipid and glucose metabolism in vivo and to induces FXR target genes in biliary carcinoma cells and HEK293 cells. However, studies are currently lacking to rationalize the molecular mechanisms of FXR modulation by prenylflavonoids. We addressed this deficiency and report the first systematic study of FXR prenylflavonoid interactions. We combined hydrogen deuterium exchange mass spectrometry (HDX-MS) with computational studies for dissecting molecular recognition and conformational impact of prenylflavonoid interactions on the ligand binding domain (LBD) of human FXR. Four prenylflavonoids were tested: xanthohumol, a prenylated chalcone, two prenylated flavonones, namely isoxanthohumol (IX) and 8-prenylnaringenin (8PN), and a semisynthetic prenylflavonoid derivative, tetrahydroxanthohumol (TX). Enhancement of the HDX protection profile data by in silico predicted models of FXR prenylflavonoid complexes resulted in mapping of the prenylflavonoid interactions within the canonical ligand binding pocket. Our findings provide a foundation for the exploration of the chemical scaffolds of prenylated chalcones and flavanones as leads for future structure activity studies of this important nuclear receptor with potential relevance for ameliorating lipid metabolic disorders associated with obesity and MetS.
Collapse
|
33
|
Hubbard TD, Murray IA, Bisson WH, Sullivan AP, Sebastian A, Perry GH, Jablonski NG, Perdew GH. Divergent Ah Receptor Ligand Selectivity during Hominin Evolution. Mol Biol Evol 2016; 33:2648-58. [PMID: 27486223 DOI: 10.1093/molbev/msw143] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
We have identified a fixed nonsynonymous sequence difference between humans (Val381; derived variant) and Neandertals (Ala381; ancestral variant) in the ligand-binding domain of the aryl hydrocarbon receptor (AHR) gene. In an exome sequence analysis of four Neandertal and Denisovan individuals compared with nine modern humans, there are only 90 total nucleotide sites genome-wide for which archaic hominins are fixed for the ancestral nonsynonymous variant and the modern humans are fixed for the derived variant. Of those sites, only 27, including Val381 in the AHR, also have no reported variability in the human dbSNP database, further suggesting that this highly conserved functional variant is a rare event. Functional analysis of the amino acid variant Ala381 within the AHR carried by Neandertals and nonhuman primates indicate enhanced polycyclic aromatic hydrocarbon (PAH) binding, DNA binding capacity, and AHR mediated transcriptional activity compared with the human AHR. Also relative to human AHR, the Neandertal AHR exhibited 150-1000 times greater sensitivity to induction of Cyp1a1 and Cyp1b1 expression by PAHs (e.g., benzo(a)pyrene). The resulting CYP1A1/CYP1B1 enzymes are responsible for PAH first pass metabolism, which can result in the generation of toxic intermediates and perhaps AHR-associated toxicities. In contrast, the human AHR retains the ancestral sensitivity observed in primates to nontoxic endogenous AHR ligands (e.g., indole, indoxyl sulfate). Our findings reveal that a functionally significant change in the AHR occurred uniquely in humans, relative to other primates, that would attenuate the response to many environmental pollutants, including chemicals present in smoke from fire use during cooking.
Collapse
Affiliation(s)
- Troy D Hubbard
- Department of Veterinary and Biomedical Sciences and Center for Molecular Toxicology and Carcinogenesis, Pennsylvania State University
| | - Iain A Murray
- Department of Veterinary and Biomedical Sciences and Center for Molecular Toxicology and Carcinogenesis, Pennsylvania State University
| | - William H Bisson
- Department of Environmental and Molecular Toxicology, Oregon State University
| | | | | | - George H Perry
- Department of Biology, Pennsylvania State University Department of Anthropology, Pennsylvania State University
| | | | - Gary H Perdew
- Department of Veterinary and Biomedical Sciences and Center for Molecular Toxicology and Carcinogenesis, Pennsylvania State University
| |
Collapse
|
34
|
Kim H, Banerjee N, Ivanov I, Pfent CM, Prudhomme KR, Bisson WH, Dashwood RH, Talcott ST, Mertens-Talcott SU. Comparison of anti-inflammatory mechanisms of mango (Mangifera Indica L.) and pomegranate (Punica Granatum L.) in a preclinical model of colitis. Mol Nutr Food Res 2016; 60:1912-23. [PMID: 27028006 DOI: 10.1002/mnfr.201501008] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 03/15/2016] [Accepted: 03/17/2016] [Indexed: 12/12/2022]
Abstract
SCOPE Tannin-rich fruits have been evaluated as alternative prevention strategies for colorectal cancer based on their anti-inflammatory properties. This study compared tannin-rich preparations from mango (rich in gallotannins) and pomegranate (rich in ellagitannins) in the dextran sodium sulfate-induced colitis model. METHODS AND RESULTS In rats, mango and pomegranate beverages decreased intestinal inflammation and the levels of pro-inflammatory cytokines in mucosa and serum. The mango beverage suppressed the ratio of phosphorylated/total protein expression of the IGF-1R-AKT/mTOR axis and downregulated mRNA expression of Igf1, Insr, and pik3cv. Pomegranate decreased p70S6K and RPS6, as well as Rps6ka2, Map2k2, and Mapk1 mRNA. In silico modeling indicated a high binding of docked of gallic acid to the catalytic domain of IGF-1R, which may suppress the activity of the enzyme. Ellagic acid docked effectively into the catalytic domains of both IGF-1R and EGFR. In vitro assays with lipopolysaccharide-treated CCD-18Co cells using polyphenolic extracts from each beverage, as well as pure compounds, corroborated the predictions made in silico. CONCLUSION Mango polyphenols inhibited the IGF-1R- AKT/mTOR axis, and pomegranate polyphenols downregulate the mTOR downstream pathway through reductions in ERK1/2. These results suggest that extracts rich in gallo- and ellagitannins act on different molecular targets in the protection against ulcerative colitis.
Collapse
Affiliation(s)
- Hyemee Kim
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, USA
| | - Nivedita Banerjee
- Interdisciplinary Program of Toxicology, Texas A&M University, College Station, TX, USA
| | - Ivan Ivanov
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, USA
| | - Catherine M Pfent
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, USA
| | - Kalan R Prudhomme
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, USA
| | - William H Bisson
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, USA
| | - Roderick H Dashwood
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, USA.,Center for Epigenetics & Disease Prevention, Texas A&M Health Science Center, Houston, TX, USA
| | - Stephen T Talcott
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, USA
| | - Susanne U Mertens-Talcott
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, USA. .,Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
35
|
Jiang C, Xie C, Lv Y, Li J, Krausz KW, Shi J, Brocker CN, Desai D, Amin SG, Bisson WH, Liu Y, Gavrilova O, Patterson AD, Gonzalez FJ. Intestine-selective farnesoid X receptor inhibition improves obesity-related metabolic dysfunction. Nat Commun 2015; 6:10166. [PMID: 26670557 PMCID: PMC4682112 DOI: 10.1038/ncomms10166] [Citation(s) in RCA: 403] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 11/10/2015] [Indexed: 12/17/2022] Open
Abstract
The farnesoid X receptor (FXR) regulates bile acid, lipid and glucose metabolism. Here we show that treatment of mice with glycine-β-muricholic acid (Gly-MCA) inhibits FXR signalling exclusively in intestine, and improves metabolic parameters in mouse models of obesity. Gly-MCA is a selective high-affinity FXR inhibitor that can be administered orally and prevents, or reverses, high-fat diet-induced and genetic obesity, insulin resistance and hepatic steatosis in mice. The high-affinity FXR agonist GW4064 blocks Gly-MCA action in the gut, and intestine-specific Fxr-null mice are unresponsive to the beneficial effects of Gly-MCA. Mechanistically, the metabolic improvements with Gly-MCA depend on reduced biosynthesis of intestinal-derived ceramides, which directly compromise beige fat thermogenic function. Consequently, ceramide treatment reverses the action of Gly-MCA in high-fat diet-induced obese mice. We further show that FXR signalling in ileum biopsies of humans positively correlates with body mass index. These data suggest that Gly-MCA may be a candidate for the treatment of metabolic disorders. The nuclear farnesoid X receptor (FXR) is activated by bile acids and influences energy metabolism. Here, the authors report a small molecule inhibitor of FXR, glycine-ß-muricholic acid, which inhibits FXR in the intestine and improves metabolic homeostasis by repressing intestinal ceramide synthesis.
Collapse
Affiliation(s)
- Changtao Jiang
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA.,Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, and the Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
| | - Cen Xie
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Ying Lv
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, and the Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
| | - Jing Li
- Department of Gastroenterology, Peking University People's Hospital, Beijing 100044, China
| | - Kristopher W Krausz
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Jingmin Shi
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Chad N Brocker
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Dhimant Desai
- Department of Pharmacology, College of Medicine, The Pennsylvania State University, Hershey, Pennsylvania 17033, USA
| | - Shantu G Amin
- Department of Pharmacology, College of Medicine, The Pennsylvania State University, Hershey, Pennsylvania 17033, USA
| | - William H Bisson
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon 97331, USA
| | - Yulan Liu
- Department of Gastroenterology, Peking University People's Hospital, Beijing 100044, China
| | - Oksana Gavrilova
- Mouse Metabolism Core Laboratory, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Andrew D Patterson
- Department of Veterinary and Biomedical Sciences and the Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
36
|
Adaptation of the human aryl hydrocarbon receptor to sense microbiota-derived indoles. Sci Rep 2015; 5:12689. [PMID: 26235394 PMCID: PMC4522678 DOI: 10.1038/srep12689] [Citation(s) in RCA: 252] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 07/06/2015] [Indexed: 02/08/2023] Open
Abstract
Ligand activation of the aryl hydrocarbon (AHR) has profound effects upon the immunological status of the gastrointestinal tract, establishing and maintaining signaling networks, which facilitate host-microbe homeostasis at the mucosal interface. However, the identity of the ligand(s) responsible for such AHR-mediated activation within the gut remains to be firmly established. Here, we combine in vitro ligand binding, quantitative gene expression, protein-DNA interaction and ligand structure activity analyses together with in silico modeling of the AHR ligand binding domain to identify indole, a microbial tryptophan metabolite, as a human-AHR selective agonist. Human AHR, acting as a host indole receptor may exhibit a unique bimolecular (2:1) binding stoichiometry not observed with typical AHR ligands. Such bimolecular indole-mediated activation of the human AHR within the gastrointestinal tract may provide a foundation for inter-kingdom signaling between the enteric microflora and the immune system to promote commensalism within the gut.
Collapse
|
37
|
Goodale BC, La Du J, Tilton SC, Sullivan CM, Bisson WH, Waters KM, Tanguay RL. Ligand-Specific Transcriptional Mechanisms Underlie Aryl Hydrocarbon Receptor-Mediated Developmental Toxicity of Oxygenated PAHs. Toxicol Sci 2015; 147:397-411. [PMID: 26141390 DOI: 10.1093/toxsci/kfv139] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Polycyclic aromatic hydrocarbons (PAHs) are priority environmental contaminants that exhibit mutagenic, carcinogenic, proinflammatory, and teratogenic properties. Oxygen-substituted PAHs (OPAHs) are formed during combustion processes and via phototoxidation and biological degradation of parent (unsubstituted) PAHs. Despite their prevalence both in contaminated industrial sites and in urban air, OPAH mechanisms of action in biological systems are relatively understudied. Like parent PAHs, OPAHs exert structure-dependent mutagenic activities and activation of the aryl hydrocarbon receptor (AHR) and cytochrome p450 metabolic pathway. Four-ring OPAHs 1,9-benz-10-anthrone (BEZO) and benz(a)anthracene-7,12-dione (7,12-B[a]AQ) cause morphological aberrations and induce markers of oxidative stress in developing zebrafish with similar potency, but only 7,12-B[a]AQ induces robust Cyp1a protein expression. We investigated the role of the AHR in mediating the toxicity of BEZO and 7,12-B[a]AQ, and found that knockdown of AHR2 rescued developmental effects caused by both compounds. Using RNA-seq and molecular docking, we identified transcriptional responses that precede developmental toxicity induced via differential interaction with AHR2. Redox-homeostasis genes were affected similarly by these OPAHs, while 7,12-B[a]AQ preferentially activated phase 1 metabolism and BEZO uniquely decreased visual system genes. Analysis of biological functions and upstream regulators suggests that BEZO is a weak AHR agonist, but interacts with other transcriptional regulators to cause developmental toxicity in an AHR-dependent manner. Identifying ligand-dependent AHR interactions and signaling pathways is essential for understanding toxicity of this class of environmentally relevant compounds.
Collapse
Affiliation(s)
- B C Goodale
- *Department of Environmental and Molecular Toxicology, The Environmental Health Sciences Center, Oregon State University, Corvallis, Oregon 97330; Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire 03755
| | - J La Du
- *Department of Environmental and Molecular Toxicology, The Environmental Health Sciences Center, Oregon State University, Corvallis, Oregon 97330
| | - S C Tilton
- *Department of Environmental and Molecular Toxicology, The Environmental Health Sciences Center, Oregon State University, Corvallis, Oregon 97330; Computational Biology and Bioinformatics, Pacific Northwest National Laboratory, Richland, Washington 99354
| | - C M Sullivan
- *Department of Environmental and Molecular Toxicology, The Environmental Health Sciences Center, Oregon State University, Corvallis, Oregon 97330; Center for Genome Research and Biocomputing, Oregon State University, Corvallis, Oregon 97330
| | - W H Bisson
- *Department of Environmental and Molecular Toxicology, The Environmental Health Sciences Center, Oregon State University, Corvallis, Oregon 97330
| | - K M Waters
- Computational Biology and Bioinformatics, Pacific Northwest National Laboratory, Richland, Washington 99354
| | - R L Tanguay
- *Department of Environmental and Molecular Toxicology, The Environmental Health Sciences Center, Oregon State University, Corvallis, Oregon 97330;
| |
Collapse
|
38
|
Anti-androgen flutamide suppresses hepatocellular carcinoma cell proliferation via the aryl hydrocarbon receptor mediated induction of transforming growth factor-β1. Oncogene 2015; 34:6092-104. [PMID: 25867062 DOI: 10.1038/onc.2015.55] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 12/07/2014] [Accepted: 12/22/2014] [Indexed: 12/17/2022]
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor and a member of the basic helix-loop-helix PER/ARNT/SIM family of chemosensors and developmental regulators. The AhR is widely known as a mediator of dioxin toxicity; however, it also suppresses cancer cell proliferation and recent findings have implicated its role as a tumor suppressor. We conducted a chemical library screen to identify nontoxic AhR ligands with anti-cancer effects and discovered flutamide (Eulexin) as a putative AhR ligand. Flutamide is an androgen receptor (AR) antagonist approved by the United States Food and Drug Administration for the treatment of prostate cancer. We found that flutamide inhibited the growth of several cancer cell lines independent of AR status, and that suppression of AhR expression reversed the anti-proliferative effects of flutamide. We investigated the AhR-dependent mechanism of action of flutamide in human hepatocellular carcinoma cells and identified that transforming growth factor-β1 (TGF-β1) is induced by flutamide in an AhR-dependent manner. In contrast, the potent AhR agonist 2,3,7,8-Tetrachlorodibenzo-p-dioxin had no effect on TGF-β1 expression, indicating the ligand specificity of AhR activation. We also determined that TGF-β1 induction is required for the AhR-dependent growth inhibitory effects of flutamide. Therefore, flutamide may be effective in AhR-positive cancers that are sensitive to TGF-β1 signaling, such as hepatocellular carcinoma.
Collapse
|
39
|
Hirano M, Hwang JH, Park HJ, Bak SM, Iwata H, Kim EY. In silico analysis of the interaction of avian aryl hydrocarbon receptors and dioxins to decipher isoform-, ligand-, and species-specific activations. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2015; 49:3795-804. [PMID: 25692546 DOI: 10.1021/es505733f] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
The aryl hydrocarbon receptor (AHR) mediates toxic responses to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and other dioxin-like compounds (DLCs). Avian species possess multiple AHR isoforms (AHR1, AHR1β, and AHR2) that exhibit species- and isoform-specific responses to ligands. To account for the ligand preference in terms of the structural features of avian AHRs, we generated in silico homology models of the ligand-binding domain of avian AHRs based on holo human HIF-2α (PDB entry 3H7W ). Molecular docking simulations of TCDD and other DLCs with avian AHR1s and AHR2s using ASEDock indicated that the interaction energy increased with the number of substituted chlorine atoms in congeners, supporting AHR transactivation potencies and World Health Organization TCDD toxic equivalency factors of congeners. The potential interaction energies of an endogenous AHR ligand, 6-formylindolo [3,2-b] carbazole (FICZ) to avian AHRs were lower than those of TCDD, which was supported by a greater potency of FICZ for in vitro AHR-mediated transactivation than TCDD. The molecular dynamics simulation revealed that mean square displacements in Ile324 and Ser380 of TCDD-bound AHR1 of the chicken, the most sensitive species to TCDD, were smaller than those in other avian AHR1s, suggesting that the dynamic stability of these amino acid residues contribute to TCDD preference. For avian AHR2, the corresponding residues (Val/Ser or Val/Ala type) were not responsible for differential TCDD sensitivity. Application of the three-dimensional reference interaction site model showed that the stabilization of TCDD binding to avian AHRs may be due to the solvation effect depending on the characteristics of two amino acids corresponding to Ile324 and Ser380 in chicken AHR1. This study demonstrates that in silico simulations of AHRs and ligands could be used to predict isoform-, ligand-, and species-specific interactions.
Collapse
Affiliation(s)
- Masashi Hirano
- †Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama 790-8577, Japan
| | - Ji-Hee Hwang
- ‡Department of Life and Nanopharmaceutical Science and Department of Biology, Kyung Hee University, Hoegi-Dong, Dongdaemun-Gu, Seoul 130-701, Korea
| | - Hae-Jeong Park
- ‡Department of Life and Nanopharmaceutical Science and Department of Biology, Kyung Hee University, Hoegi-Dong, Dongdaemun-Gu, Seoul 130-701, Korea
| | - Su-Min Bak
- ‡Department of Life and Nanopharmaceutical Science and Department of Biology, Kyung Hee University, Hoegi-Dong, Dongdaemun-Gu, Seoul 130-701, Korea
| | - Hisato Iwata
- †Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama 790-8577, Japan
| | - Eun-Young Kim
- ‡Department of Life and Nanopharmaceutical Science and Department of Biology, Kyung Hee University, Hoegi-Dong, Dongdaemun-Gu, Seoul 130-701, Korea
| |
Collapse
|
40
|
Fratev F. Activation helix orientation of the estrogen receptor is mediated by receptor dimerization: evidence from molecular dynamics simulations. Phys Chem Chem Phys 2015; 17:13403-20. [DOI: 10.1039/c5cp00327j] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
ERα dimer formation reshapes the helix 12 conformational landscape and is a leading factor for the activation helix conformation.
Collapse
Affiliation(s)
- Filip Fratev
- Institute of Biophysics and Biomedical Engineering
- Bulgarian Academy of Sciences
- 1113 Sofia
- Bulgaria
- Micar21 Ltd
| |
Collapse
|