1
|
Sahoo R, Pattnaik S, Mohanty B, Mir SA, Behera B. Aryl hydrocarbon receptor (AHR) signalling: A double-edged sword guiding both cancer progression and cancer therapy. Biochim Biophys Acta Gen Subj 2025; 1869:130805. [PMID: 40222634 DOI: 10.1016/j.bbagen.2025.130805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/21/2025] [Accepted: 04/05/2025] [Indexed: 04/15/2025]
Abstract
Aryl Hydrocarbon Receptor (AHR) reported to be associated with major carcinogenic signalling cascades which cause cell proliferations, metastasis and invasion as well as immune imbalance. AHR Participates in cellular processes not only through genomic pathways to cause genomic alterations but also via nongenomic pathways to alter various cytoplasmic proteins. In addition, AHR senses a wide range of ligands that modulate its downstream mechanisms that are intricated in cancer induction and prevention. Thus, AHR functions as a two-sided sword where some AHR ligands contribute to enhance cancer whereas few are useful for cancer treatment. Therefore, AHR represent as a regulatory point in cancer progression and treatment. There is a need to reinvestigate the regulatory role of AHR in major intracellular pathways and to explore the potential of AHR ligand for the design of cancer therapeutics. This review emphasizes the interaction of AHR with pro-carcinogenic signalling pathways that modulate cancer induction and progression. Furthermore, it also discusses about the current discovery of AHR ligands for cancer initiation or inhibition. This information could be useful for development of therapeutic strategies for the management of cancer by targeting AHR.
Collapse
Affiliation(s)
- Rahul Sahoo
- Department of Biotechnology and Bioinformatics, Sambalpur University, Jyoti Vihar, Burla, Odisha 768019, India
| | - Sriya Pattnaik
- Department of Biotechnology and Bioinformatics, Sambalpur University, Jyoti Vihar, Burla, Odisha 768019, India
| | - Biswajit Mohanty
- Department of Biotechnology and Bioinformatics, Sambalpur University, Jyoti Vihar, Burla, Odisha 768019, India
| | - Showkat Ahmad Mir
- School of Life Sciences, Sambalpur University, Jyoti Vihar, Burla, Odisha 768019, India
| | - Birendra Behera
- Department of Biotechnology and Bioinformatics, Sambalpur University, Jyoti Vihar, Burla, Odisha 768019, India.
| |
Collapse
|
2
|
Partsch V, Crudo F, Piller D, Varga E, Del Favero G, Marko D. Resolving complexity: Identification of altersetin and toxin mixtures responsible for the immunomodulatory, antiestrogenic and genotoxic potential of a complex Alternaria mycotoxin extract. Food Chem Toxicol 2025; 198:115315. [PMID: 39933689 DOI: 10.1016/j.fct.2025.115315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/29/2025] [Accepted: 02/07/2025] [Indexed: 02/13/2025]
Abstract
Alternaria mycotoxins may pose significant risks to human health due to their diverse spectrum of adverse effects and frequent occurrences in food. A previous study demonstrated the immunosuppressive, antiestrogenic, and genotoxic potential of a complex Alternaria mycotoxin extract (CE). The present study aimed to elucidate specific Alternaria mycotoxins or combinations thereof responsible for toxicity. Following toxicity-guided fractionation of the CE, a multiparametric panel of assays was applied to assess different endpoints. These included immunomodulatory effects (NF-κB reporter gene assay in THP1-Lucia™ monocytes), estrogenicity/antiestrogenicity (alkaline phosphatase assay in Ishikawa cells) and genotoxicity (γH2AX and alkaline comet assays in HepG2 cells). LC-MS/MS analysis revealed prominent mycotoxins in the active fractions, with altersetin (AST) identified as a novel key compound exhibiting immunoinhibitory (≥2 μM) and antiestrogenic (≥5 μM) properties in vitro. Additionally, while specific mycotoxin combinations explained the toxicity of active fractions, some effects remained unexplained, suggesting the presence of unidentified bioactive substances. This study underscores the significance of AST and specific toxin mixtures as major contributors to CE toxicity. Further, it highlights the importance of considering combinatory effects in risk assessment of Alternaria mycotoxins as well as further investigation of unknown Alternaria metabolites, which may pose additional health risks.
Collapse
Affiliation(s)
- Vanessa Partsch
- University of Vienna, Faculty of Chemistry, Department of Food Chemistry and Toxicology, 1090, Vienna, Austria; University of Vienna, Faculty of Chemistry, Doctoral School in Chemistry, 1090, Vienna, Austria
| | - Francesco Crudo
- University of Vienna, Faculty of Chemistry, Department of Food Chemistry and Toxicology, 1090, Vienna, Austria.
| | - Daniel Piller
- University of Vienna, Faculty of Chemistry, Department of Food Chemistry and Toxicology, 1090, Vienna, Austria
| | - Elisabeth Varga
- University of Vienna, Faculty of Chemistry, Department of Food Chemistry and Toxicology, 1090, Vienna, Austria; University of Veterinary Medicine Vienna, Clinical Department for Farm Animals and Food System Science, Centre for Food Science and Veterinary Public Health, Unit Food Hygiene and Technology, 1210, Vienna, Austria
| | - Giorgia Del Favero
- University of Vienna, Faculty of Chemistry, Department of Food Chemistry and Toxicology, 1090, Vienna, Austria; University of Vienna, Faculty of Chemistry, Core Facility Multimodal Imaging, 1090, Vienna, Austria
| | - Doris Marko
- University of Vienna, Faculty of Chemistry, Department of Food Chemistry and Toxicology, 1090, Vienna, Austria
| |
Collapse
|
3
|
Qiu S, Ding X, Ma X, Zhang L, Chen J, Wei W. Muscle cells affect the promoting effect of FGF21 on lipid accumulation in porcine adipocytes through AhR/FGFR1 signaling pathway. Biochem Biophys Res Commun 2025; 754:151520. [PMID: 40015071 DOI: 10.1016/j.bbrc.2025.151520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/01/2025]
Abstract
The intramuscular fat (IMF) content, as an important meat quality trait, can directly affect the tenderness, juiciness, and flavor of pork. Reasonably increasing the IMF content can improve the palatability of pork. Therefore, identification of important factors for the lipid accumulation among muscles is the breakthrough point for improving meat quality. FGF21, identified as a novel metabolic regulator, has been found to regulate glucose and lipid metabolism in 3T3-L1 adipocytes, but its function in porcine adipocytes remains unclear. In this study, we discovered that the administration of recombinant FGF21 protein promotes adipogenic differentiation and increases triglyceride accumulation in porcine adipocytes. While the expression of FGFR1 in adipocytes under muscle conditions is inhibited, affecting the signal transduction of FGF21. This inhibitory effect is accompanied by activation of the AhR signaling pathway. When treated with the AhR antagonist CH223191, there was a partial restoration of FGFR1 expression levels. This indicates that muscle cells suppress the expression of FGFR1 in adipocytes by activating the AhR signaling pathway, thereby affecting the signal transduction of FGF21. Our results reveal the regulatory role of FGF21 in pig adipocyte differentiation and the regulatory mechanism of muscle environment on FGFR1 expression, providing new theoretical basis for IMF content improvement from the perspective of FGF21-FGFR1 signaling transduction.
Collapse
Affiliation(s)
- Shengda Qiu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Xiaolei Ding
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Xiangfei Ma
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Lifan Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Jie Chen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Wei Wei
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
4
|
Olafsen NE, Das S, Gorrini C, Matthews J. Long-term exposure to BAY2416964 reduces proliferation, migration and recapitulates transcriptional changes induced by AHR loss in PyMT-induced mammary tumor cells. Front Oncol 2024; 14:1466658. [PMID: 39450255 PMCID: PMC11499230 DOI: 10.3389/fonc.2024.1466658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024] Open
Abstract
The aryl hydrocarbon receptor (AHR) is a ligand activated transcription factor which in certain cancer types drives pro-survival processes that facilitate tumorigenesis, malignant cell migration, invasion, and metastasis. Much of AHR's pro-tumorigenic action is due to its activation by the oncometabolite, kynurenine. Because of this AHR antagonists are being actively investigated as new anti-tumor therapy. In this study we compared the effects of treatment with the AHR antagonists, BAY2416964 and GNF351, to that of AHR knockout in PyMT murine mammary cancer cells. BAY2416964 and GNF351 effectively inhibited kynurenine-dependent increases in Cyp1a1 and Cyp1b1 mRNA levels. CRISPR/Cas9-generated PyMT AhrKO cells exhibited reduced cell proliferation compared with controls, but treatment with 1 μM BAY2416964 for 96 h had no effect on the proliferation of wildtype cells. To further examine the differences between AHR knockout and short term BAY2416964, we generated long-term BAY2416964 (LT-BAY) cells by exposing wildtype cells to 1 μM BAY2416964 for at least 6 weeks. Similar to AhrKO cells, LT-BAY cells exhibited reduced cell proliferation and migration compared with wildtype cells. No differentially expressed genes (DEGs) were identified in wildtype cells exposed to 1 μM BAY2416964 for 24 h; however, 46.4% of DEGs overlapped between AhrKO and LT-BAY cells including gene regulated cell proliferation. Our data reveal long-term pharmacological inhibition of AHR by BAY2416964 closely resembles AHR loss in a mouse model of breast cancer.
Collapse
Affiliation(s)
- Ninni Elise Olafsen
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Siddhartha Das
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Chiara Gorrini
- School of Molecular and Cellular Biology, University of Leeds, Leeds, United Kingdom
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Jason Matthews
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
5
|
Mosa FES, Alqahtani MA, El-Ghiaty MA, Dyck JRB, Barakat K, El-Kadi AOS. Identification of aryl hydrocarbon receptor allosteric antagonists from clinically approved drugs. Drug Dev Res 2024; 85:e22232. [PMID: 38992915 DOI: 10.1002/ddr.22232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 06/14/2024] [Accepted: 06/30/2024] [Indexed: 07/13/2024]
Abstract
The human aryl hydrocarbon receptor (AhR), a ligand-dependent transcription factor, plays a pivotal role in a diverse array of pathways in biological and pathophysiological events. This position AhR as a promising target for both carcinogenesis and antitumor strategies. In this study we utilized computational modeling to screen and identify FDA-approved drugs binding to the allosteric site between α2 of bHLH and PAS-A domains of AhR, with the aim of inhibiting its canonical pathway activity. Our findings indicated that nilotinib effectively fits into the allosteric pocket and forms interactions with crucial residues F82, Y76, and Y137. Binding free energy value of nilotinib is the lowest among top hits and maintains stable within its pocket throughout entire (MD) simulations time. Nilotinib has also substantial interactions with F295 and Q383 when it binds to orthosteric site and activate AhR. Surprisingly, it does not influence AhR nuclear translocation in the presence of AhR agonists; instead, it hinders the formation of the functional AhR-ARNT-DNA heterodimer assembly, preventing the upregulation of regulated enzymes like CYP1A1. Importantly, nilotinib exhibits a dual impact on AhR, modulating AhR activity via the PAS-B domain and working as a noncompetitive allosteric antagonist capable of blocking the canonical AhR signaling pathway in the presence of potent AhR agonists. These findings open a new avenue for the repositioning of nilotinib beyond its current application in diverse diseases mediated via AhR.
Collapse
Affiliation(s)
- Farag E S Mosa
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Mohammed A Alqahtani
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Mahmoud A El-Ghiaty
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Jason R B Dyck
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Khaled Barakat
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Ayman O S El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
6
|
Houser CL, Fenner KN, Lawrence BP. Timing influences the impact of aryl hydrocarbon receptor activation on the humoral immune response to respiratory viral infection. Toxicol Appl Pharmacol 2024; 489:117010. [PMID: 38901696 PMCID: PMC11240840 DOI: 10.1016/j.taap.2024.117010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/05/2024] [Accepted: 06/15/2024] [Indexed: 06/22/2024]
Abstract
Humoral responses to respiratory viruses, such as influenza viruses, develop over time and are central to protection from repeated infection with the same or similar viruses. Epidemiological and experimental studies have linked exposures to environmental contaminants that bind the aryl hydrocarbon receptor (AHR) with modulated antibody responses to pathogenic microorganisms and common vaccinations. Other studies have prompted investigation into the potential therapeutic applications of compounds that activate AHR. Herein, using two different AHR ligands [2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and 2-(1H-Indol-3-ylcarbonyl)-4-thiazolecarboxylic acid methyl ester (ITE), to modulate the duration of AHR activity, we show that the humoral response to viral infection is dependent upon the duration and timing of AHR signaling, and that different cellular elements of the response have different sensitivities. When AHR activation was initiated prior to infection with influenza A virus, there was suppression of all measured elements of the humoral response (i.e., the frequency of T follicular helper cells, germinal center B cells, plasma cells, and circulating virus-specific antibody). However, when the timing of AHR activation was adjusted to either early (days -1 to +5 relative to infection) or later (days +5 onwards), then AHR activation affected different aspects of the overall humoral response. These findings highlight the importance of considering the timing of AHR activation in relation to triggering an immune response, particularly when targeting the AHR to manipulate disease processes.
Collapse
Affiliation(s)
- Cassandra L Houser
- Department of Microbiology & Immunology, University of Rochester, Rochester NY14642, USA
| | - Kristina N Fenner
- Department of Environmental Medicine, University of Rochester, Rochester NY14642, USA
| | - B Paige Lawrence
- Department of Microbiology & Immunology, University of Rochester, Rochester NY14642, USA; Department of Environmental Medicine, University of Rochester, Rochester NY14642, USA.
| |
Collapse
|
7
|
Tian Y, Rimal B, Bisanz JE, Gui W, Wolfe TM, Koo I, Murray IA, Nettleford SK, Yokoyama S, Dong F, Koshkin S, Prabhu KS, Turnbaugh PJ, Walk ST, Perdew GH, Patterson AD. Effects of Early Life Exposures to the Aryl Hydrocarbon Receptor Ligand TCDF on Gut Microbiota and Host Metabolic Homeostasis in C57BL/6J Mice. ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:87005. [PMID: 39140734 PMCID: PMC11323762 DOI: 10.1289/ehp13356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 04/30/2024] [Accepted: 07/08/2024] [Indexed: 08/15/2024]
Abstract
BACKGROUND Exposure to persistent organic pollutants (POPs) and disruptions in the gastrointestinal microbiota have been positively correlated with a predisposition to factors such as obesity, metabolic syndrome, and type 2 diabetes; however, it is unclear how the microbiome contributes to this relationship. OBJECTIVE This study aimed to explore the association between early life exposure to a potent aryl hydrocarbon receptor (AHR) agonist and persistent disruptions in the microbiota, leading to impaired metabolic homeostasis later in life. METHODS This study used metagenomics, nuclear magnetic resonance (NMR)- and mass spectrometry (MS)-based metabolomics, and biochemical assays to analyze the gut microbiome composition and function, as well as the physiological and metabolic effects of early life exposure to 2,3,7,8-tetrachlorodibenzofuran (TCDF) in conventional, germ-free (GF), and Ahr-null mice. The impact of TCDF on Akkermansia muciniphila (A. muciniphila) in vitro was assessed using optical density (OD 600), flow cytometry, transcriptomics, and MS-based metabolomics. RESULTS TCDF-exposed mice exhibited lower abundances of A. muciniphila, lower levels of cecal short-chain fatty acids (SCFAs) and indole-3-lactic acid (ILA), as well as lower levels of the gut hormones glucagon-like peptide 1 (GLP-1) and peptide YY (PYY), findings suggestive of disruption in the gut microbiome community structure and function. Importantly, microbial and metabolic phenotypes associated with early life POP exposure were transferable to GF recipients in the absence of POP carry-over. In addition, AHR-independent interactions between POPs and the microbiota were observed, and they were significantly associated with growth, physiology, gene expression, and metabolic activity outcomes of A. muciniphila, supporting suppressed activity along the ILA pathway. CONCLUSIONS These data obtained in a mouse model point to the complex effects of POPs on the host and microbiota, providing strong evidence that early life, short-term, and self-limiting POP exposure can adversely impact the microbiome, with effects persisting into later life with associated health implications. https://doi.org/10.1289/EHP13356.
Collapse
Affiliation(s)
- Yuan Tian
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University (Penn State), University Park, Pennsylvania, USA
- Huck Institutes of the Life Sciences, Penn State, University Park, Pennsylvania, USA
| | - Bipin Rimal
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University (Penn State), University Park, Pennsylvania, USA
| | - Jordan E. Bisanz
- Department of Biochemistry and Molecular Biology, Penn State, University Park, Pennsylvania, USA
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, California, USA
| | - Wei Gui
- Huck Institutes of the Life Sciences, Penn State, University Park, Pennsylvania, USA
| | - Trenton M. Wolfe
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, USA
| | - Imhoi Koo
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University (Penn State), University Park, Pennsylvania, USA
| | - Iain A. Murray
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University (Penn State), University Park, Pennsylvania, USA
| | - Shaneice K. Nettleford
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University (Penn State), University Park, Pennsylvania, USA
| | - Shigetoshi Yokoyama
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University (Penn State), University Park, Pennsylvania, USA
| | - Fangcong Dong
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University (Penn State), University Park, Pennsylvania, USA
| | - Sergei Koshkin
- Huck Institutes of the Life Sciences, Penn State, University Park, Pennsylvania, USA
| | - K. Sandeep Prabhu
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University (Penn State), University Park, Pennsylvania, USA
| | - Peter J. Turnbaugh
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, California, USA
- Chan Zuckerberg Biohub–San Francisco, San Francisco, California, USA
| | - Seth T. Walk
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, USA
| | - Gary H. Perdew
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University (Penn State), University Park, Pennsylvania, USA
| | - Andrew D. Patterson
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University (Penn State), University Park, Pennsylvania, USA
| |
Collapse
|
8
|
Heine S, Alessandrini F, Grosch J, Graß C, Heldner A, Schnautz B, Grosch J, Buters J, Slusarenko BO, Krappmann D, Fallarino F, Ohnmacht C, Schmidt-Weber CB, Blank S. Activation of the aryl hydrocarbon receptor improves allergen-specific immunotherapy of murine allergic airway inflammation: a novel adjuvant option? Front Immunol 2024; 15:1397072. [PMID: 38915403 PMCID: PMC11194380 DOI: 10.3389/fimmu.2024.1397072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 05/29/2024] [Indexed: 06/26/2024] Open
Abstract
Background Allergen-specific immunotherapy (AIT) is able to restore immune tolerance to allergens in allergic patients. However, some patients do not or only poorly respond to current treatment protocols. Therefore, there is a need for deeper mechanistic insights and further improvement of treatment strategies. The relevance of the aryl hydrocarbon receptor (AhR), a ligand-dependent transcription factor, has been investigated in several inflammatory diseases, including allergic asthma. However, its potential role in AIT still needs to be addressed. Methods A murine model of AIT in ovalbumin-induced allergic airway inflammation was performed in AhR-deficient (AhR-/-) and wild-type mice. Furthermore, AIT was combined with the application of the high-affinity AhR agonist 10-chloro-7H-benzimidazo[2,1-a]benzo[de]iso-quinolin-7-one (10-Cl-BBQ) as an adjuvant to investigate the effects of AhR activation on therapeutic outcome. Results Although AhR-/- mice suffer stronger allergic responses than wild-type mice, experimental AIT is comparably effective in both. Nevertheless, combining AIT with the administration of 10-Cl-BBQ improved therapeutic effects by an AhR-dependent mechanism, resulting in decreased cell counts in the bronchoalveolar fluid, decreased pulmonary Th2 and Th17 cell levels, and lower sIgE levels. Conclusion This study demonstrates that the success of AIT is not dependent on the AhR. However, targeting the AhR during AIT can help to dampen inflammation and improve tolerogenic vaccination. Therefore, AhR ligands might represent promising candidates as immunomodulators to enhance the efficacy of AIT.
Collapse
Affiliation(s)
- Sonja Heine
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Health & Helmholtz Munich, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Munich, Germany
| | - Francesca Alessandrini
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Health & Helmholtz Munich, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Munich, Germany
| | - Johannes Grosch
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Health & Helmholtz Munich, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Munich, Germany
| | - Carina Graß
- Research Unit Signaling and Translation, Group Signaling and Immunity, Molecular Targets and Therapeutic Center, Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
| | - Alexander Heldner
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Health & Helmholtz Munich, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Munich, Germany
| | - Benjamin Schnautz
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Health & Helmholtz Munich, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Munich, Germany
| | - Johanna Grosch
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Health & Helmholtz Munich, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Munich, Germany
| | - Jeroen Buters
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Health & Helmholtz Munich, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Munich, Germany
| | - Benjamin O. Slusarenko
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Health & Helmholtz Munich, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Munich, Germany
| | - Daniel Krappmann
- Research Unit Signaling and Translation, Group Signaling and Immunity, Molecular Targets and Therapeutic Center, Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
| | | | - Caspar Ohnmacht
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Health & Helmholtz Munich, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Munich, Germany
| | - Carsten B. Schmidt-Weber
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Health & Helmholtz Munich, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Munich, Germany
| | - Simon Blank
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Health & Helmholtz Munich, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Munich, Germany
| |
Collapse
|
9
|
Gripshover TC, Wahlang B, Head KZ, Luo J, Bolatimi OE, Smith ML, Rouchka EC, Chariker JH, Xu J, Cai L, Cummins TD, Merchant ML, Zheng H, Kong M, Cave MC. Multiomics Analysis of PCB126's Effect on a Mouse Chronic-Binge Alcohol Feeding Model. ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:47007. [PMID: 38619879 PMCID: PMC11018247 DOI: 10.1289/ehp14132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 04/17/2024]
Abstract
BACKGROUND Environmental pollutants, including polychlorinated biphenyls (PCBs) have been implicated in the pathogenesis of liver disease. Our group recently demonstrated that PCB126 promoted steatosis, hepatomegaly, and modulated intermediary metabolism in a rodent model of alcohol-associated liver disease (ALD). OBJECTIVE To better understand how PCB126 promoted ALD in our previous model, the current study adopts multiple omics approaches to elucidate potential mechanistic hypotheses. METHODS Briefly, male C57BL/6J mice were exposed to 0.2 mg / kg polychlorinated biphenyl (PCB) 126 or corn oil vehicle prior to ethanol (EtOH) or control diet feeding in the chronic-binge alcohol feeding model. Liver tissues were collected and prepared for mRNA sequencing, phosphoproteomics, and inductively coupled plasma mass spectrometry for metals quantification. RESULTS Principal component analysis showed that PCB126 uniquely modified the transcriptome in EtOH-fed mice. EtOH feeding alone resulted in > 4,000 differentially expressed genes (DEGs), and PCB126 exposure resulted in more DEGs in the EtOH-fed group (907 DEGs) in comparison with the pair-fed group (503 DEGs). Top 20 significant gene ontology (GO) biological processes included "peptidyl tyrosine modifications," whereas top 25 significantly decreasing GO molecular functions included "metal/ion/zinc binding." Quantitative, label-free phosphoproteomics and western blot analysis revealed no major significant PCB126 effects on total phosphorylated tyrosine residues in EtOH-fed mice. Quantified hepatic essential metal levels were primarily significantly lower in EtOH-fed mice. PCB126-exposed mice had significantly lower magnesium, cobalt, and zinc levels in EtOH-fed mice. DISCUSSION Previous work has demonstrated that PCB126 is a modifying factor in metabolic dysfunction-associated steatotic liver disease (MASLD), and our current work suggests that pollutants also modify ALD. PCB126 may, in part, be contributing to the malnutrition aspect of ALD, where metal deficiency is known to contribute and worsen prognosis. https://doi.org/10.1289/EHP14132.
Collapse
Affiliation(s)
- Tyler C. Gripshover
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, School of Medicine, University of Louisville, Louisville, Kentucky, USA
- University of Louisville Superfund Research Program, University of Louisville, Louisville, Kentucky, USA
| | - Banrida Wahlang
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, School of Medicine, University of Louisville, Louisville, Kentucky, USA
- University of Louisville Superfund Research Program, University of Louisville, Louisville, Kentucky, USA
- The Center for Integrative Environmental Health Sciences, University of Louisville, Louisville, Kentucky, USA
| | - Kimberly Z. Head
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, School of Medicine, University of Louisville, Louisville, Kentucky, USA
- Hepatobiology & Toxicology COBRE, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Jianzhu Luo
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Oluwanifemi E. Bolatimi
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Melissa L. Smith
- The Center for Integrative Environmental Health Sciences, University of Louisville, Louisville, Kentucky, USA
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Eric C. Rouchka
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Kentucky IDeA Network of Biomedical Research Excellence (KY INBRE) Bioinformatics Core, University of Louisville, Louisville, Kentucky, USA
| | - Julia H. Chariker
- Kentucky IDeA Network of Biomedical Research Excellence (KY INBRE) Bioinformatics Core, University of Louisville, Louisville, Kentucky, USA
- Department of Neuroscience Training, University of Louisville, Louisville, Kentucky, USA
| | - Jason Xu
- Department of Pediatrics, Pediatric Research Institute, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Lu Cai
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, Kentucky, USA
- The Center for Integrative Environmental Health Sciences, University of Louisville, Louisville, Kentucky, USA
- Department of Pediatrics, Pediatric Research Institute, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Timothy D. Cummins
- Division of Nephrology and Hypertension, Department of Medicine and Clinical Proteomics Center, University of Louisville, Louisville, Kentucky, USA
| | - Michael L. Merchant
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, Kentucky, USA
- The Center for Integrative Environmental Health Sciences, University of Louisville, Louisville, Kentucky, USA
- Division of Nephrology and Hypertension, Department of Medicine and Clinical Proteomics Center, University of Louisville, Louisville, Kentucky, USA
- University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky, USA
| | - Hao Zheng
- The Center for Integrative Environmental Health Sciences, University of Louisville, Louisville, Kentucky, USA
| | - Maiying Kong
- The Center for Integrative Environmental Health Sciences, University of Louisville, Louisville, Kentucky, USA
- Department of Bioinformatics and Biostatistics School of Public Health and Information Sciences, University of Louisville, Louisville, Kentucky, USA
- University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky, USA
- Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | - Matthew C. Cave
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, School of Medicine, University of Louisville, Louisville, Kentucky, USA
- University of Louisville Superfund Research Program, University of Louisville, Louisville, Kentucky, USA
- The Center for Integrative Environmental Health Sciences, University of Louisville, Louisville, Kentucky, USA
- Hepatobiology & Toxicology COBRE, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, Kentucky, USA
- University of Louisville Alcohol Research Center, University of Louisville, Louisville, Kentucky, USA
- The Robley Rex Veterans Affairs Medical Center, Louisville, Kentucky, USA
- The Liver Transplant Program at UofL Health – Jewish Hospital Trager Transplant Center, Louisville, Kentucky, USA
| |
Collapse
|
10
|
Li K, Li K, He Y, Liang S, Shui X, Lei W. Aryl hydrocarbon receptor: A bridge linking immuno-inflammation and metabolism in atherosclerosis. Biochem Pharmacol 2023; 216:115744. [PMID: 37579858 DOI: 10.1016/j.bcp.2023.115744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 08/16/2023]
Abstract
Cardiovascular disease is the leading cause of death worldwide, and atherosclerosis is a major contributor to this etiology. The ligand-activated transcription factor, known as the aryl hydrocarbon receptor (AhR), plays an essential role in the interactions between genes and the environment. In a number of human diseases, including atherosclerosis, the AhR signaling pathway has recently been shown to be aberrantly expressed and activated. It's reported that AhR can regulate the immuno-inflammatory response and metabolism pathways in atherosclerosis, potentially serving as a bridge that links these processes. In this review, we highlight the involvement of AhR in atherosclerosis. From the literature, we conclude that AhR is a potential target for controlling atherosclerosis through precise interventions.
Collapse
Affiliation(s)
- Kongwei Li
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Department of Precision Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Kaiyue Li
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Department of Precision Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yuan He
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Shan Liang
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Department of Precision Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Xiaorong Shui
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Laboratory of Vascular Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China.
| | - Wei Lei
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Department of Precision Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China.
| |
Collapse
|
11
|
Haghshenas R, Aftabi Y, Doaei S, Gholamalizadeh M. Synergistic effect of endurance training and nettle leaf extract on the IDO1-KYN-AHR pathway homeostasis and inhibiting of liver toxicity in rats with STZ-induced diabetes. Front Endocrinol (Lausanne) 2023; 14:1071424. [PMID: 37305057 PMCID: PMC10251405 DOI: 10.3389/fendo.2023.1071424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 05/05/2023] [Indexed: 06/13/2023] Open
Abstract
Introduction Diabetes adversely affects a number of hepatic molecular pathways, including the kynurenine (KYN) pathway. KYN is produced by indoleamine 2,3-dioxygenase (IDO) and activates the aryl hydrocarbon receptor (AHR). This study evaluated the effect of endurance training (EndTr) and nettle leaf extract (NLE) on the IDO1-KYN-AHR pathway in the livers of rats with streptozotocin-induced diabetes. Methods We divided 48 rats into six groups: controls (Ct), treated with EndTr (EndTr), diabetes-induced (D), D treated with NLE (D + NLE), D treated with EndTr (D + EnTr), and D treated with EndTr and NLE (D + EndTr + NLE). EndTr, D + EnTr, and D + EndTr + NLE groups were subjected to training with running on treadmill for 8 weeks, 5 days per week, 25 min in first session to 59 min at last session with intensity of 55% to 65% VO2max. Using real-time PCR gene (Ahr, Cyp1a1, and Ido1) expressions and ELISA, malondialdehyde (MDA) and protein (IDO1, AHR, and CYP1A1) levels were determined in the liver samples. Results A significant three-way interaction of exercise, nettle, and diabetes was observed on the all variables (P< 0.001). In particular, significant increases in blood glucose level (BGL), in gene and protein expression, and in MDA and KYN levels were observed in the liver samples of the D group versus the Ct group (P< 0.05). BGL and liver MDA levels were significantly lower in the D + EndTr and D + NLE groups than that in the D group. However, the D + EndTr + NLE group showed a more significant decrease in these factors (P< 0.05). In addition, liver KYN levels were significantly lower in the EndTr group compared with that in the Ct group as well as in the D + EndTr + NLE and D + EndTr groups compared with that in the D groups (P< 0.05). Whereas both the EndTr and D + NLE groups showed lower Ahr expression and AHR level compared with the Ct and D groups, respectively (P< 0.05), the D + EndTr + NLE group showed a higher significant reduction in the AHR level than the D group (P< 0.05). The Cyp1a1 expression and IDO1 level significantly decreased only in the D + EndTr + NLE group compared to that in the D group (P< 0.05). Conclusion Overall, this study showed that the combination of EndTr and NLE may synergistically restore the imbalanced IDO1-KYN-AHR pathway in diabetic liver.
Collapse
Affiliation(s)
- Rouhollah Haghshenas
- Department of Sport Sciences, Faculty of Humanities, Semnan University, Semnan, Iran
| | - Younes Aftabi
- Tuberculosis and Lung Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saied Doaei
- Department of Community Nutrition, School of Nutrition and Food Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Gholamalizadeh
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Giordano D, Facchiano A, Moccia S, Meola AMI, Russo GL, Spagnuolo C. Molecular Docking of Natural Compounds for Potential Inhibition of AhR. Foods 2023; 12:foods12101953. [PMID: 37238771 DOI: 10.3390/foods12101953] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 05/04/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
The aryl hydrocarbon receptor (AhR) is a highly conserved environmental sensor, historically known for mediating the toxicity of xenobiotics. It is involved in numerous cellular processes such as differentiation, proliferation, immunity, inflammation, homeostasis, and metabolism. It exerts a central role in several conditions such as cancer, inflammation, and aging, acting as a transcription factor belonging to the basic helix-loop-helix/Per-ARNT-Sim (bHLH-PAS) protein family. A key step in the canonical AhR activation is AhR-ARNT heterodimerization followed by the binding to the xenobiotic-responsive elements (XREs). The present work aims to investigate the potential AhR inhibitory activity of selected natural compounds. Due to the absence of a complete structure of human AhRs, a model consisting of the bHLH, the PAS A, and the PAS B domains was constructed. Blind and focused docking simulations revealed the presence of further binding pockets, different from the canonical one presented in the PAS B domain, which could be important for AhR inhibition due to the possibility to impede AhR:ARNT heterodimerization, either preventing conformational changes or masking crucial sites necessary for protein-protein interaction. Two of the compounds retrieved from the docking simulations, i.e., β-carotene and ellagic acid, confirmed their capacity of inhibiting benzo[a]pyrene (BaP)-induced AhR activation in in vitro tests on the human hepatoma cell line HepG2, validating the efficacy of the computational approach.
Collapse
Affiliation(s)
- Deborah Giordano
- National Research Council, Institute of Food Sciences, 83100 Avellino, Italy
| | - Angelo Facchiano
- National Research Council, Institute of Food Sciences, 83100 Avellino, Italy
| | - Stefania Moccia
- National Research Council, Institute of Food Sciences, 83100 Avellino, Italy
| | | | - Gian Luigi Russo
- National Research Council, Institute of Food Sciences, 83100 Avellino, Italy
| | - Carmela Spagnuolo
- National Research Council, Institute of Food Sciences, 83100 Avellino, Italy
| |
Collapse
|
13
|
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
|
14
|
Pedroni L, Louisse J, Dorne JLCM, Dall'Asta C, Dellafiora L. A computational study on the biotransformation of alkenylbenzenes by a selection of CYPs: Reflections on their possible bioactivation. Toxicology 2023; 488:153471. [PMID: 36863505 DOI: 10.1016/j.tox.2023.153471] [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: 01/25/2023] [Revised: 02/23/2023] [Accepted: 02/27/2023] [Indexed: 03/03/2023]
Abstract
Alkenylbenzenes are aromatic compounds found in several vegetable foods that can cause genotoxicity upon bioactivation by members of the cytochrome P450 (CYP) family, forming 1'-hydroxy metabolites. These intermediates act as proximate carcinogens and can be further converted into reactive 1'-sulfooxy metabolites, which are the ultimate carcinogens responsible for genotoxicity. Safrole, a member of this class, has been banned as a food or feed additive in many countries based on its genotoxicity and carcinogenicity. However, it can still enter the food and feed chain. There is limited information about the toxicity of other alkenylbenzenes that may be present in safrole-containing foods, such as myristicin, apiole, and dillapiole. In vitro studies showed safrole as mainly bioactivated by CYP2A6 to form its proximate carcinogen, while for myristicin this is mainly done by CYP1A1. However, it is not known whether CYP1A1 and CYP2A6 can activate apiole and dillapiole. The present study uses an in silico pipeline to investigate this knowledge gap and determine whether CYP1A1 and CYP2A6 may play a role in the bioactivation of these alkenylbenzenes. The study found that the bioactivation of apiole and dillapiole by CYP1A1 and CYP2A6 is limited, possibly indicating that these compounds may have limited toxicity, while describing a possible role of CYP1A1 in the bioactivation of safrole. The study expands the current understanding of safrole toxicity and bioactivation and helps understand the mechanisms of CYPs involved in the bioactivation of alkenylbenzenes. This information is essential for a more informed analysis of alkenylbenzenes toxicity and risk assessment.
Collapse
Affiliation(s)
- Lorenzo Pedroni
- Department of Food and Drug, University of Parma, Parma 43124, Italy
| | - Jochem Louisse
- Wageningen Food Safety Research, P.O. Box 230, 6700 AE Wageningen, the Netherlands
| | | | - Chiara Dall'Asta
- Department of Food and Drug, University of Parma, Parma 43124, Italy
| | - Luca Dellafiora
- Department of Food and Drug, University of Parma, Parma 43124, Italy.
| |
Collapse
|
15
|
Yucel MA, Ozcelik I, Algul O. Machine learning study: from the toxicity studies to tetrahydrocannabinol effects on Parkinson's disease. Future Med Chem 2023; 15:365-377. [PMID: 36942739 DOI: 10.4155/fmc-2022-0181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023] Open
Abstract
Aim: Investigating molecules having toxicity and chemical similarity to find hit molecules. Methods: The machine learning (ML) model was developed to predict the arylhydrocarbon receptor (AHR) activity of anti-Parkinson's and US FDA-approved drugs. The ML algorithm was a support vector machine, and the dataset was Tox21. Results: The ML model predicted apomorphine in anti-Parkinson's drugs and 73 molecules in FDA-approved drugs as active. The authors were curious if there is any molecule like apomorphine in these 73 molecules. A fingerprint similarity analysis of these molecules was conducted and found tetrahydrocannabinol (THC). Molecular docking studies of THC for dopamine receptor 1 (affinity = -8.2 kcal/mol) were performed. Conclusion: THC may affect dopamine receptors directly and could be useful for Parkinson's disease.
Collapse
Affiliation(s)
- Mehmet Ali Yucel
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Erzincan Binali Yildirim University, Erzincan, 24100, Turkey
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Mersin University, Mersin, 33169, Turkey
| | - Ibrahim Ozcelik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Erzincan Binali Yildirim University, Erzincan, 24100, Turkey
| | - Oztekin Algul
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Erzincan Binali Yildirim University, Erzincan, 24100, Turkey
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Mersin University, Mersin, 33169, Turkey
| |
Collapse
|
16
|
Zhao L, Yao L, Chen R, He J, Lin T, Qiu S, Chen G, Chen H, Qiu SX. Pinostrobin from plants and propolis against human coronavirus HCoV-OC43 by modulating host AHR/CYP1A1 pathway and lipid metabolism. Antiviral Res 2023; 212:105570. [PMID: 36863496 PMCID: PMC9974210 DOI: 10.1016/j.antiviral.2023.105570] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 02/10/2023] [Accepted: 02/21/2023] [Indexed: 03/04/2023]
Abstract
Coronaviruses, as enveloped positive-strand RNA viruses, manipulate host lipid compositions to enable robust viral replication. Temporal modulation of the host lipid metabolism is a potential novel strategy against coronaviruses. Here, the dihydroxyflavone pinostrobin (PSB) was identified through bioassay that inhibited the increment of human coronavirus OC43 (HCoV-OC43) in human ileocecal colorectal adenocarcinoma cells. Lipid metabolomic studies showed that PSB interfered with linoleic acid and arachidonic acid metabolism pathways. PSB significantly decreased the level of 12, 13- epoxyoctadecenoic (12, 13-EpOME) and increased the level of prostaglandin E2. Interestingly, exogenous supplement of 12, 13-EpOME in HCoV-OC43-infected cells significantly stimulated HCoV-OC43 virus replication. Transcriptomic analyses showed that PSB is a negative modulator of aryl hydrocarbon receptor (AHR)/cytochrome P450 (CYP) 1A1signaling pathway and its antiviral effects can be counteracted by supplement of FICZ, a well-known AHR agonist. Integrative analyses of metabolomic and transcriptomic indicated that PSB could affect linoleic acid and arachidonic acid metabolism axis through AHR/CYP1A1 pathway. These results highlight the importance of the AHR/CYP1A1 pathway and lipid metabolism in the anti-coronavirus activity of the bioflavonoid PSB.
Collapse
Affiliation(s)
- Liyun Zhao
- Key Laboratory of Plant Resources Conservation and Sustainable Utilization, Guangdong Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, 510650, PR China
| | - Liyuan Yao
- Key Laboratory of Plant Resources Conservation and Sustainable Utilization, Guangdong Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, 510650, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Rui Chen
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Jiani He
- Key Laboratory of Plant Resources Conservation and Sustainable Utilization, Guangdong Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, 510650, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Tingting Lin
- Key Laboratory of Plant Resources Conservation and Sustainable Utilization, Guangdong Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, 510650, PR China
| | - Silin Qiu
- Key Laboratory of Plant Resources Conservation and Sustainable Utilization, Guangdong Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, 510650, PR China
| | - Guohua Chen
- Key Laboratory of Plant Resources Conservation and Sustainable Utilization, Guangdong Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, 510650, PR China
| | - Hongfeng Chen
- Key Laboratory of Plant Resources Conservation and Sustainable Utilization, Guangdong Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, 510650, PR China
| | - Sheng-Xiang Qiu
- Key Laboratory of Plant Resources Conservation and Sustainable Utilization, Guangdong Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, 510650, PR China.
| |
Collapse
|
17
|
Tseng CY, Custer CM, Custer TW, Dummer PM, Karouna-Renier N, Matson CW. Multi-omics responses in tree swallow (Tachycineta bicolor) nestlings from the Maumee Area of Concern, Maumee River, Ohio. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 856:159130. [PMID: 36183771 DOI: 10.1016/j.scitotenv.2022.159130] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 09/26/2022] [Accepted: 09/26/2022] [Indexed: 06/16/2023]
Abstract
A multi-omics approach was utilized to identify altered biological responses and functions, and to prioritize contaminants to assess the risks of chemical mixtures in the Maumee Area of Concern (AOC), Maumee River, OH, USA. The Maumee AOC is designated by the United States Environmental Protection Agency as having significant beneficial use impairments, including degradation of fish and wildlife populations, bird or animal deformities or reproduction problems, and loss of fish and wildlife habitat. Tree swallow (Tachycineta bicolor) nestlings were collected at five sites along the Maumee River, which included wastewater treatment plants (WWTPs) and industrial land-use sites. Polychlorinated biphenyls (PCBs), polybrominated diphenyl ethers (PBDEs), polycyclic aromatic hydrocarbons (PAHs), polychlorinated dibenzo p dioxins and furans (PCDD/Fs), and chlorinated pesticide concentrations were elevated in Maumee tree swallows, relative to a remote reference site, Star Lake, WI, USA. Liver tissue was utilized for non-targeted transcriptome and targeted metabolome evaluation. A significantly differentially expressed gene cluster related to a downregulation in cell growth and cell cycle regulation was identified when comparing all Maumee River sites with the reference site. There was an upregulation of lipogenesis genes, such as PPAR signaling (HMGCS2, SLC22A5), biosynthesis of unsaturated fatty acids (FASN, SCD, ELOVL2, and FADS2), and higher lipogenesis related metabolites, such as docosapentaenoic acid (DPA), docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA), and arachidonic acid (AA) at two industrial land-use sites, Ironhead and Maumee, relative to WWTP sites (Perrysburg and SideCut), and the reference site. Toledo Water, in the vicinity of the other two industrial sites and also adjacent to a WWTP, showed a mix of signals between industrial land-use and WWTP land-use. PAHs, oxychlordane, and PBDEs were determined to be the most likely causes of the differentiation in biological responses, including de novo lipogenesis and biosynthesis of unsaturated fatty acids.
Collapse
Affiliation(s)
- Chi Yen Tseng
- Department of Environmental Science, The Institute of Ecological, Earth, and Environmental Sciences (TIE3S), the Center for Reservoir and Aquatic Systems Research (CRASR), Baylor University, Waco, TX 76798, United States
| | - Christine M Custer
- Upper Midwest Environmental Sciences Center, U.S. Geological Survey, La Crosse, WI 54603, United States
| | - Thomas W Custer
- Upper Midwest Environmental Sciences Center, U.S. Geological Survey, La Crosse, WI 54603, United States
| | - Paul M Dummer
- Upper Midwest Environmental Sciences Center, U.S. Geological Survey, La Crosse, WI 54603, United States
| | - Natalie Karouna-Renier
- U.S. Geological Survey, Eastern Ecological Science Center (EESC) at Patuxent, Beltsville, MD 20705, United States
| | - Cole W Matson
- Department of Environmental Science, The Institute of Ecological, Earth, and Environmental Sciences (TIE3S), the Center for Reservoir and Aquatic Systems Research (CRASR), Baylor University, Waco, TX 76798, United States.
| |
Collapse
|
18
|
Eccles KM, Karmaus AL, Kleinstreuer NC, Parham F, Rider CV, Wambaugh JF, Messier KP. A geospatial modeling approach to quantifying the risk of exposure to environmental chemical mixtures via a common molecular target. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 855:158905. [PMID: 36152849 PMCID: PMC9979101 DOI: 10.1016/j.scitotenv.2022.158905] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/09/2022] [Accepted: 09/17/2022] [Indexed: 05/14/2023]
Abstract
In the real world, individuals are exposed to chemicals from sources that vary over space and time. However, traditional risk assessments based on in vivo animal studies typically use a chemical-by-chemical approach and apical disease endpoints. New approach methodologies (NAMs) in toxicology, such as in vitro high-throughput (HTS) assays generated in Tox21 and ToxCast, can more readily provide mechanistic chemical hazard information for chemicals with no existing data than in vivo methods. In this paper, we establish a workflow to assess the joint action of 41 modeled ambient chemical exposures in the air from the USA-wide National Air Toxics Assessment by integrating human exposures with hazard data from curated HTS (cHTS) assays to identify counties where exposure to the local chemical mixture may perturb a common biological target. We exemplify this proof-of-concept using CYP1A1 mRNA up-regulation. We first estimate internal exposure and then convert the inhaled concentration to a steady state plasma concentration using physiologically based toxicokinetic modeling parameterized with county-specific information on ages and body weights. We then use the estimated blood plasma concentration and the concentration-response curve from the in vitro cHTS assay to determine the chemical-specific effects of the mixture components. Three mixture modeling methods were used to estimate the joint effect from exposure to the chemical mixture on the activity levels, which were geospatially mapped. Finally, a Monte Carlo uncertainty analysis was performed to quantify the influence of each parameter on the combined effects. This workflow demonstrates how NAMs can be used to predict early-stage biological perturbations that can lead to adverse health outcomes that result from exposure to chemical mixtures. As a result, this work will advance mixture risk assessment and other early events in the effects of chemicals.
Collapse
Affiliation(s)
- Kristin M Eccles
- National Institute of Environmental Health Science, Division of the Translational Toxicology, Durham, USA
| | - Agnes L Karmaus
- Integrated Laboratory Systems, an Inotiv Company, Morrisville, NC, USA
| | - Nicole C Kleinstreuer
- National Institute of Environmental Health Science, Division of the Translational Toxicology, Durham, USA
| | - Fred Parham
- National Institute of Environmental Health Science, Division of the Translational Toxicology, Durham, USA
| | - Cynthia V Rider
- National Institute of Environmental Health Science, Division of the Translational Toxicology, Durham, USA
| | - John F Wambaugh
- United States Environmental Protection Agency, Center for Computational Toxicology and Exposure, Durham, USA
| | - Kyle P Messier
- National Institute of Environmental Health Science, Division of the Translational Toxicology, Durham, USA.
| |
Collapse
|
19
|
Skóra B, Matuszewska P, Masicz M, Sikora K, Słomczewska M, Sołtysek P, Szychowski KA. Crosstalk between the aryl hydrocarbon receptor (AhR) and the peroxisome proliferator-activated receptor gamma (PPARγ) as a key factor in the metabolism of silver nanoparticles in neuroblastoma (SH-SY5Y) cells in vitro. Toxicol Appl Pharmacol 2023; 458:116339. [PMID: 36473513 DOI: 10.1016/j.taap.2022.116339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/20/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022]
Abstract
The potential usefulness of silver nanoparticles (AgNPs) in anticancer therapy has been postulated for many years. However, little is known to date about the exact impact of such NPs on intracellular detoxication pathways. Therefore, the aim of this study was to determine the impact of AgNPs on the AhR-PPARγ-CYP1A1 pathway in neuroblastoma (SH-SY5Y) cells. The obtained results showed a decrease in the metabolic activity of the SH-SY5Y cells at the 50 and 100 μg/mL concentrations with an increase in caspase-3 activity. An increase in the intercellular ROS production was observed at the 1 and 10 μg/mL concentrations. The co-treatment of the AgNP-treated cells with the AhR and PPARγ inhibitors abolished the effect of the tested AgNPs in the SH-SY5Y cells. In turn, the CYP1A1 activity assay showed a decrease in this parameter in the AgNP-treated cells. Moreover, the gene expression analysis demonstrated that AgNPs were able to increase the AhR and CYP1A1 mRNA expression and decrease the PPARγ gene expression after the 6-h treatment. In turn, an increase in the AhR and PPARγ protein expression was observed after 24 h. Summarizing, the study shows for the first time that AgNPs with a 5-nm diameter size are able to exert a cytotoxic effect on SH-SH5Y cells in a ROS-dependent manner affect the AhR-PPARγ-CYP1A1 pathway inter alia by inhibiting the activity of CYP1A1. This is important due to given present research approaches using such NPs as enhancer agents in the modern PPARγ inhibitor-based anticancer therapy.
Collapse
Affiliation(s)
- Bartosz Skóra
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management, St. Sucharskiego 2, 35-225 Rzeszow, Poland.
| | - Paulina Matuszewska
- Medical Biotechnology Student's Science Group "Helisa", Medical College, University of Information Technology and Management, St. Sucharskiego 2, 35-225 Rzeszow, Poland
| | - Martyna Masicz
- Medical Biotechnology Student's Science Group "Helisa", Medical College, University of Information Technology and Management, St. Sucharskiego 2, 35-225 Rzeszow, Poland
| | - Karolina Sikora
- Medical Biotechnology Student's Science Group "Helisa", Medical College, University of Information Technology and Management, St. Sucharskiego 2, 35-225 Rzeszow, Poland
| | - Magnolia Słomczewska
- Medical Biotechnology Student's Science Group "Helisa", Medical College, University of Information Technology and Management, St. Sucharskiego 2, 35-225 Rzeszow, Poland
| | - Paulina Sołtysek
- Medical Biotechnology Student's Science Group "Helisa", Medical College, University of Information Technology and Management, St. Sucharskiego 2, 35-225 Rzeszow, Poland
| | - Konrad A Szychowski
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management, St. Sucharskiego 2, 35-225 Rzeszow, Poland
| |
Collapse
|
20
|
Tian X, Zheng S, Wang J, Yu M, Lin Z, Qin M, Wu Y, Chen S, Zhong S. Cardiac disorder-related adverse events for aryl hydrocarbon receptor agonists: a safety review. Expert Opin Drug Saf 2022; 21:1505-1510. [PMID: 35582860 DOI: 10.1080/14740338.2022.2078301] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Although cardiac disorder-related adverse events (AEs) have been reported in patients treated with aryl hydrocarbon receptor (AHR) agonists, their safety profiles remain unknown. Here, we identified significant cardiac disorders associated with AHR agonists and further evaluated their relevance. RESEARCH DESIGN AND METHODS Database queries were performed using OpenVigil 2.1 and AEs voluntarily submitted to Food and Drug Administration Adverse Event Reporting System (FAERS) between 2004 and 2020 were included. This study based on the Medical Dictionary for Regulatory Activities and the standardized MedDRA Queries to define the preferred terms, and we used reporting odd ratio to detect signals. RESULTS In the FAERS database, 14,078 cardiac disorder-related AEs were identified in patients receiving AHR agonists. Among all AHR agonists, the number of cardiac disorder-related PTs with positive signals for AHR agonists was 93. Peripheral swelling (n = 1572) and atrial fibrillation (n = 1277) were the most reported cardiac disorder-related AEs among AHR agonists in disproportionately reported PTs. Moreover, several AHR agonists were highly associated with tachyarrhythmia. CONCLUSIONS By mining the FAERS database, we provided more information on the association between AHR agonist use and cardiac disorder-related AEs.
Collapse
Affiliation(s)
- Xiaoxue Tian
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Shufen Zheng
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Jing Wang
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Meiling Yu
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhuoheng Lin
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Min Qin
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuanyuan Wu
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Shiyu Chen
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Shilong Zhong
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China.,Department of Cardiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| |
Collapse
|
21
|
Iizuka T, Yin P, Zuberi A, Kujawa S, Coon JS, Björvang RD, Damdimopoulou P, Pacyga DC, Strakovsky RS, Flaws JA, Bulun SE. Mono-(2-ethyl-5-hydroxyhexyl) phthalate promotes uterine leiomyoma cell survival through tryptophan-kynurenine-AHR pathway activation. Proc Natl Acad Sci U S A 2022; 119:e2208886119. [PMID: 36375056 PMCID: PMC9704719 DOI: 10.1073/pnas.2208886119] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 10/03/2022] [Indexed: 11/15/2022] Open
Abstract
Uterine leiomyoma is the most common tumor in women and causes severe morbidity in 15 to 30% of reproductive-age women. Epidemiological studies consistently indicate a correlation between leiomyoma development and exposure to endocrine-disrupting chemical phthalates, especially di-(2-ethylhexyl) phthalate (DEHP); however, the underlying mechanisms are unknown. Here, among the most commonly encountered phthalate metabolites, we found the strongest association between the urine levels of mono(2-ethyl-5-hydroxyhexyl) phthalate (MEHHP), the principal DEHP metabolite, and the risk of uterine leiomyoma diagnosis (n = 712 patients). The treatment of primary leiomyoma and smooth muscle cells (n = 29) with various mixtures of phthalate metabolites, at concentrations equivalent to those detected in urine samples, significantly increased cell viability and decreased apoptosis. MEHHP had the strongest effects on both cell viability and apoptosis. MEHHP increased cellular tryptophan and kynurenine levels strikingly and induced the expression of the tryptophan transporters SLC7A5 and SLC7A8, as well as, tryptophan 2,3-dioxygenase (TDO2), the key enzyme catalyzing the conversion of tryptophan to kynurenine that is the endogenous ligand of aryl hydrocarbon receptor (AHR). MEHHP stimulated nuclear localization of AHR and up-regulated the expression of CYP1A1 and CYP1B1, two prototype targets of AHR. siRNA knockdown or pharmacological inhibition of SLC7A5/SLC7A8, TDO2, or AHR abolished MEHHP-mediated effects on leiomyoma cell survival. These findings indicate that MEHHP promotes leiomyoma cell survival by activating the tryptophan-kynurenine-AHR pathway. This study pinpoints MEHHP exposure as a high-risk factor for leiomyoma growth, uncovers a mechanism by which exposure to environmental phthalate impacts leiomyoma pathogenesis, and may lead to the development of novel druggable targets.
Collapse
Affiliation(s)
- Takashi Iizuka
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60610
| | - Ping Yin
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60610
| | - Azna Zuberi
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60610
| | - Stacy Kujawa
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60610
| | - John S. Coon
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60610
| | - Richelle D. Björvang
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institute and Karolinska University Hospital, 171 64 Stockholm, Sweden
| | - Pauliina Damdimopoulou
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institute and Karolinska University Hospital, 171 64 Stockholm, Sweden
| | - Diana C. Pacyga
- Department of Food Science and Human Nutrition, Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824
| | - Rita S. Strakovsky
- Department of Food Science and Human Nutrition, Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824
| | - Jodi A. Flaws
- Department of Comparative Bioscience, University of Illinois at Urbana–Champagne, Urbana, IL 61802
| | - Serdar E. Bulun
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60610
| |
Collapse
|
22
|
Safe S, Zhang L. The Role of the Aryl Hydrocarbon Receptor (AhR) and Its Ligands in Breast Cancer. Cancers (Basel) 2022; 14:5574. [PMID: 36428667 PMCID: PMC9688153 DOI: 10.3390/cancers14225574] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 10/27/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
Breast cancer is a complex disease which is defined by numerous cellular and molecular markers that can be used to develop more targeted and successful therapies. The aryl hydrocarbon receptor (AhR) is overexpressed in many breast tumor sub-types, including estrogen receptor -positive (ER+) tumors; however, the prognostic value of the AhR for breast cancer patient survival is not consistent between studies. Moreover, the functional role of the AhR in various breast cancer cell lines is also variable and exhibits both tumor promoter- and tumor suppressor- like activity and the AhR is expressed in both ER-positive and ER-negative cells/tumors. There is strong evidence demonstrating inhibitory AhR-Rα crosstalk where various AhR ligands induce ER degradation. It has also been reported that different structural classes of AhR ligands, including halogenated aromatics, polynuclear aromatics, synthetic drugs and other pharmaceuticals, health promoting phytochemical-derived natural products and endogenous AhR-active compounds inhibit one or more of breast cancer cell proliferation, survival, migration/invasion, and metastasis. AhR-dependent mechanisms for the inhibition of breast cancer by AhR agonists are variable and include the downregulation of multiple genes/gene products such as CXCR4, MMPs, CXCL12, SOX4 and the modulation of microRNA levels. Some AhR ligands, such as aminoflavone, have been investigated in clinical trials for their anticancer activity against breast cancer. In contrast, several publications have reported that AhR agonists and antagonists enhance and inhibit mammary carcinogenesis, respectively, and differences between the anticancer activities of AhR agonists in breast cancer may be due in part to cell context and ligand structure. However, there are reports showing that the same AhR ligand in the same breast cancer cell line gives opposite results. These differences need to be resolved in order to further develop and take advantage of promising agents that inhibit mammary carcinogenesis by targeting the AhR.
Collapse
Affiliation(s)
- Stephen Safe
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | | |
Collapse
|
23
|
Modulation of Aryl Hydrocarbon Receptor Expression Alleviated Neuropathic Pain in a Chronic Constriction Nerve Injury Animal Model. Int J Mol Sci 2022; 23:ijms231911255. [PMID: 36232555 PMCID: PMC9570158 DOI: 10.3390/ijms231911255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/17/2022] [Accepted: 09/19/2022] [Indexed: 12/02/2022] Open
Abstract
Neuropathic pain is well known to occur after damage to the somatosensory system. Aryl hydrocarbon receptor (AhR) has neuroprotective effects when the central nervous system is subjected to internal and external stimulations. However, the exact mechanism by which AhR regulates neuropathic pain is poorly understood. Nerve explant culture and the chronic constrictive nerve injury (CCI) model in wild or AhR-knockout mice were used in this study. In the nerve explant culture, the ovoid number increased in the AhR−/− condition and was decreased by omeprazole (AhR agonist) in a dose-dependent manner. Increased nerve degeneration and the associated inflammation response appeared in the AhR−/− condition, and these changes were attenuated by omeprazole. High expression of AhR in the injured nerve was noted after CCI. Deletion of AhR aggravated nerve damages and this was restored by omeprazole. Deletion of AhR increased NGF expression and reduced axon number in the paw skin, but this was attenuated by omeprazole. A highly expressed inflammation reaction over the dorsal spinal cord, somatosensory cortex, and hippocampus was noted in the AhR-deleted animals. Administration of omeprazole attenuated not only the inflammatory response, but also the amplitude of somatosensory evoked potential. Deletion of AhR further aggravated the neurobehavior compared with the wild type, but such behavior was attenuated by omeprazole. Chronic constrictive nerve injury augmented AhR expression of the injured nerve, and AhR deletion worsened the damage, while AhR agonist omeprazole counteracted such changes. AhR agonists could be potential candidates for neuropathic pain treatment.
Collapse
|
24
|
Yoda T, Tochitani T, Usui T, Kouchi M, Inada H, Hosaka T, Kanno Y, Miyawaki I, Yoshinari K. Involvement of the CYP1A1 inhibition-mediated activation of aryl hydrocarbon receptor in drug-induced hepatotoxicity. J Toxicol Sci 2022; 47:359-373. [PMID: 36047110 DOI: 10.2131/jts.47.359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Hepatotoxicity is one of the most common toxicities observed in non-clinical safety studies of drug candidates, and it is important to understand the hepatotoxicity mechanism to assess the risk of drug-induced liver injury in humans. In this study, we investigated the mechanism of hepatotoxicity caused by 2-[2-Methyl-1-(oxan-4-yl)-1H-benzimidazol-5-yl]-1,3-benzoxazole (DSP-0640), a drug candidate that showed hepatotoxicity characterized by centrilobular hypertrophy and vacuolation of hepatocytes in a 4-week oral repeated-dose toxicity study in male rats. In the liver of rats treated with DSP-0640, the expression of aryl hydrocarbon receptor (AHR) target genes, including Cyp1a1, was upregulated. In in vitro reporter assays, however, DSP-0640 showed only minimal AHR-activating potency. Therefore, we investigated the possibility that DSP-0640 indirectly activated AHR by inhibiting the CYP1 enzyme-dependent clearance of endogenous AHR agonists. In in vitro assays, DSP-0640 showed inhibitory effects on both rat and human CYP1A1 and enhanced rat and human AHR-mediated reporter gene expression induced by 6-formylindolo[3,2-b]carbazole, a well-known endogenous AHR agonist. The possible involvement of CYP1A1 inhibition in AHR activation was also demonstrated with other hepatotoxic compounds tacrine and albendazole. These results suggest that CYP1A1 inhibition-mediated AHR activation is involved in the hepatotoxicity caused by DSP-0640 and that DSP-0640 might induce hepatotoxicity in humans as well. We propose that CYP1A1 inhibition-mediated AHR activation is a novel mechanism for drug-induced hepatotoxicity.
Collapse
Affiliation(s)
- Tomomi Yoda
- Preclinical Research Unit, Sumitomo Pharma Co., Ltd.,Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka
| | | | - Toru Usui
- Preclinical Research Unit, Sumitomo Pharma Co., Ltd
| | - Mami Kouchi
- Preclinical Research Unit, Sumitomo Pharma Co., Ltd
| | | | - Takuomi Hosaka
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka
| | - Yuichiro Kanno
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka
| | | | - Kouichi Yoshinari
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka
| |
Collapse
|
25
|
Wilson LB, McClure RS, Waters KM, Simonich MT, Tanguay RL. Concentration-response gene expression analysis in zebrafish reveals phenotypically-anchored transcriptional responses to retene. FRONTIERS IN TOXICOLOGY 2022; 4:950503. [PMID: 36093370 PMCID: PMC9453431 DOI: 10.3389/ftox.2022.950503] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/12/2022] [Indexed: 11/13/2022] Open
Abstract
Polycyclic aromatic hydrocarbons (PAHs) are ubiquitous environmental contaminants and are associated with human disease. Canonically, many PAHs induce toxicity via activation of the aryl hydrocarbon receptor (AHR) pathway. While the interaction between PAHs and the AHR is well-established, understanding which AHR-regulated transcriptional effects directly result in observable phenotypes and which are adaptive or benign is important to better understand PAH toxicity. Retene is a frequently detected PAH in environmental sampling and has been associated with AHR2-dependent developmental toxicity in zebrafish, though its mechanism of toxicity has not been fully elucidated. To interrogate transcriptional changes causally associated with retene toxicity, we conducted whole-animal RNA sequencing at 48 h post-fertilization after exposure to eight retene concentrations. We aimed to identify the most sensitive transcriptomic responses and to determine whether this approach could uncover gene sets uniquely differentially expressed at concentrations which induce a phenotype. We identified a concentration-response relationship for differential gene expression in both number of differentially expressed genes (DEGs) and magnitude of expression change. Elevated expression of cyp1a at retene concentrations below the threshold for teratogenicity suggested that while cyp1a expression is a sensitive biomarker of AHR activation, it may be too sensitive to serve as a biomarker of teratogenicity. Genes differentially expressed at only non-teratogenic concentrations were enriched for transforming growth factor-β (TGF-β) signaling pathway disruption while DEGs identified at only teratogenic concentrations were significantly enriched for response to xenobiotic stimulus and reduction-oxidation reaction activity. DEGs which spanned both non-teratogenic and teratogenic concentrations showed similar disrupted biological processes to those unique to teratogenic concentrations, indicating these processes were disrupted at low exposure concentrations. Gene co-expression network analysis identified several gene modules, including those associated with PAHs and AHR2 activation. One, Module 7, was strongly enriched for AHR2-associated genes and contained the strongest responses to retene. Benchmark concentration (BMC) of Module seven genes identified a median BMC of 7.5 µM, nearly the highest retene concentration with no associated teratogenicity, supporting the hypothesis that Module seven genes are largely responsible for retene toxicity.
Collapse
Affiliation(s)
- Lindsay B. Wilson
- Department of Environmental and Molecular Toxicology, Sinnhuber Aquatic Research Laboratory, Oregon State University, Corvallis, OR, United States
| | - Ryan S. McClure
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, United States
| | - Katrina M. Waters
- Department of Environmental and Molecular Toxicology, Sinnhuber Aquatic Research Laboratory, Oregon State University, Corvallis, OR, United States
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, United States
| | - Michael T. Simonich
- Department of Environmental and Molecular Toxicology, Sinnhuber Aquatic Research Laboratory, Oregon State University, Corvallis, OR, United States
| | - Robyn L. Tanguay
- Department of Environmental and Molecular Toxicology, Sinnhuber Aquatic Research Laboratory, Oregon State University, Corvallis, OR, United States
- *Correspondence: Robyn L. Tanguay,
| |
Collapse
|
26
|
Kynurenine Pathway-An Underestimated Factor Modulating Innate Immunity in Sepsis-Induced Acute Kidney Injury? Cells 2022; 11:cells11162604. [PMID: 36010680 PMCID: PMC9406744 DOI: 10.3390/cells11162604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/17/2022] [Accepted: 08/17/2022] [Indexed: 11/17/2022] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection, and it accounts for about half of the cases of acute kidney injury (AKI). Although sepsis is the most frequent cause of AKI in critically ill patients, its pathophysiological mechanisms are not well understood. Sepsis has the ability to modulate the function of cells belonging to the innate immune system. Increased activity of indoleamine 2,3-dioxygenase 1 (IDO1) and production of kynurenines are the major metabolic pathways utilized by innate immunity cells to maintain immunological tolerance. The activation of the kynurenine pathway (KP) plays a dual role in sepsis—in the early stage, the induction of IDO1 elicits strong proinflammatory effects that may lead to tissue damage and septic shock. Afterwards, depletion of tryptophan and production of kynurenines contribute to the development of immunosuppression that may cause the inability to overpower opportunistic infections. The presented review provides available data on the various interdependencies between elements of innate immunity and sepsis-induced AKI (SAKI) with particular emphasis on the immunomodulatory significance of KP in the above processes. We believe that KP activation may be one of the crucial, though underestimated, components of a deregulated host response to infection during SAKI.
Collapse
|
27
|
Coe KJ, Feinstein M, Higgins JW, Leung P, Scott BP, Skaptason J, Tam Y, Volak LP, Kinong J, Bittner A, McAllister H, Lim NM, Hack M, Koudriakova T. Characterization of JNJ-2482272 [4-(4-Methyl-2-(4-(Trifluoromethyl)Phenyl)Thiazole-5-yl) Pyrimidine-2-Amine] As a Strong Aryl Hydrocarbon Receptor Activator in Rat and Human. Drug Metab Dispos 2022; 50:1064-1076. [PMID: 35680134 DOI: 10.1124/dmd.121.000825] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 05/18/2022] [Indexed: 11/22/2022] Open
Abstract
[4-(4-Methyl-2-(4-(trifluoromethyl)phenyl)thiazole-5-yl)pyrimidine-2-amine] (JNJ-2482272), under investigation as an anti-inflammatory agent, was orally administered to rats once daily at 60 mg/kg for 6 consecutive days. Despite high plasma exposure after single administration (Cmax of 7.1 μM), JNJ-2482272 had plasma concentrations beneath the lower limit of quantification (3 ng/ml) after 6 consecutive days of dosing. To determine if JNJ-2482272 is an autoinducer in rats, plated rat hepatocytes were treated with JNJ-2482272 for 2 days. The major hydroxylated metabolites of JNJ-2482272 were isolated and characterized by mass spectrometry and NMR analyses. Compared with the vehicle-treated cells, a concentration-dependent increase was observed in the formation of phase I- and II-mediated metabolites coinciding with greater expression of cytochrome P450s (P450s) and UDP-glucuronosyltransferases (UGTs) in rat hepatocytes. CYP1A1, CYP1A2, CYP1B1, and UGT1A6 transcripts were predominantly induced, suggesting that JNJ-2482272 is an activator of the aryl hydrocarbon receptor (AhR). In a human AhR reporter assay, JNJ-2482272 demonstrated potent AhR activation with an EC50 value of 0.768 nM, a potency more comparable to the strong AhR activator and toxin 2,3,7,8-tetrachloro-dibenzodioxin than to weaker AhR activators 3-methylcholanthrene, β-naphthoflavone, and omeprazole. In plated human hepatocytes, JNJ-2482272 induced CYP1A1 gene expression with an EC50 of 20.4 nM and increased CYP1A activity >50-fold from basal levels. In human recombinant P450s, JNJ-2482272 was exclusively metabolized by the CYP1 family of enzymes and most rapidly by CYP1A1. The summation of these in vitro findings bridges the in vivo conclusion that JNJ-2482272 is a strong autoinducer in rats and potentially in humans through potent AhR activation. SIGNIFICANCE STATEMENT: Drugs that induce their own metabolism (autoinducers) can lack sustained exposures for pharmacology and safety assessment hindering their development. JNJ-2482272 is demonstrated herein as a strong aryl hydrocarbon receptor (AhR) activator and CYP1A autoinducer, explaining its near complete loss of exposure after repeat administration in rat, which is likely translatable to human (if progressed further) considering its nanomolar potency comparable to "classical" AhR ligands like 2,3,7,8-tetrachloro-dibenzo-dioxin despite bearing a "nonclassical" drug structure.
Collapse
Affiliation(s)
- Kevin J Coe
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Mark Feinstein
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - J William Higgins
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Perry Leung
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Brian P Scott
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Judy Skaptason
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Yuen Tam
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Laurie P Volak
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Jennifer Kinong
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Anton Bittner
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Heather McAllister
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Nathan M Lim
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Michael Hack
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Tatiana Koudriakova
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| |
Collapse
|
28
|
Development and Applications of a Zebrafish (Danio rerio) CYP1A-Targeted Monoclonal Antibody (CRC4) with Reactivity across Vertebrate Taxa: Evidence for a Conserved CYP1A Epitope. TOXICS 2022; 10:toxics10070404. [PMID: 35878309 PMCID: PMC9320060 DOI: 10.3390/toxics10070404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/17/2022] [Accepted: 07/18/2022] [Indexed: 02/06/2023]
Abstract
CYP1A is a heme-thiolate enzyme associated with the cytochrome P4501A1 monooxygenase system and is inducible by a wide variety of xenobiotics and endogenous ligands that bind and activate the aryl hydrocarbon receptor (AHR). The AHR-CYP1A axis is important for detoxification of certain xenobiotics and for homeostatic balance of endogenous sex hormones, amine hormones, vitamins, fatty acids, and phospholipids. Herein, we generated and described applications of a zebrafish CYP1A-targeted monoclonal antibody (mAb CRC4) that fortuitously recognizes induced CYP1A across vertebrate taxa, including fish, chicken, mouse, rat, and human. We then demonstrated that mAb CRC4 targets a highly conserved epitope signature of vertebrate CYP1A. The unique complimentary determining region (CDR) sequences of heavy and light chains were determined, and these Ig sequences will allow for the expression of recombinant mAb CRC4, thus superseding the need for long-term hybridoma maintenance. This antibody works well for immunohistochemistry (IHC), as well as whole-mounted IHC in zebrafish embryos. Monoclonal antibody CRC4 may be particularly useful for studying the AHR-CYP1A axis in multiple vertebrate species and within the context of Oceans and Human Health research. By using archived samples, when possible, we actively promoted efforts to reduce, replace, and refine studies involving live animals.
Collapse
|
29
|
Corton JC, Mitchell CA, Auerbach S, Bushel P, Ellinger-Ziegelbauer H, Escobar PA, Froetschl R, Harrill AH, Johnson K, Klaunig JE, Pandiri AR, Podtelezhnikov AA, Rager JE, Tanis KQ, van der Laan JW, Vespa A, Yauk CL, Pettit SD, Sistare FD. A Collaborative Initiative to Establish Genomic Biomarkers for Assessing Tumorigenic Potential to Reduce Reliance on Conventional Rodent Carcinogenicity Studies. Toxicol Sci 2022; 188:4-16. [PMID: 35404422 PMCID: PMC9238304 DOI: 10.1093/toxsci/kfac041] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
There is growing recognition across broad sectors of the scientific community that use of genomic biomarkers has the potential to reduce the need for conventional rodent carcinogenicity studies of industrial chemicals, agrochemicals, and pharmaceuticals through a weight-of-evidence approach. These biomarkers fall into 2 major categories: (1) sets of gene transcripts that can identify distinct tumorigenic mechanisms of action; and (2) cancer driver gene mutations indicative of rapidly expanding growth-advantaged clonal cell populations. This call-to-action article describes a collaborative approach launched to develop and qualify biomarker gene expression panels that measure widely accepted molecular pathways linked to tumorigenesis and their activation levels to predict tumorigenic doses of chemicals from short-term exposures. Growing evidence suggests that application of such biomarker panels in short-term exposure rodent studies can identify both tumorigenic hazard and tumorigenic activation levels for chemical-induced carcinogenicity. In the future, this approach will be expanded to include methodologies examining mutations in key cancer driver gene mutation hotspots as biomarkers of both genotoxic and nongenotoxic chemical tumor risk. Analytical, technical, and biological validation studies of these complementary genomic tools are being undertaken by multisector and multidisciplinary collaborative teams within the Health and Environmental Sciences Institute. Success from these efforts will facilitate the transition from current heavy reliance on conventional 2-year rodent carcinogenicity studies to more rapid animal- and resource-sparing approaches for mechanism-based carcinogenicity evaluation supporting internal and regulatory decision-making.
Collapse
Affiliation(s)
- J Christopher Corton
- Center for Computational Toxicology and Exposure, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Constance A Mitchell
- Health and Environmental Sciences Institute, Washington, District of Columbia, USA
| | - Scott Auerbach
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Pierre Bushel
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Durham, North Carolina, USA
| | | | - Patricia A Escobar
- Safety Assessment and Laboratory Animal Resources, Merck Sharp & Dohme Corp, West Point, Pennsylvania, USA
| | - Roland Froetschl
- BfArM-Bundesinstitut für Arzneimittel und Medizinprodukte, Federal Institute for Drugs and Medical Devices, Bonn, Germany
| | - Alison H Harrill
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | | | - James E Klaunig
- Laboratory of Investigative Toxicology and Pathology, Department of Environmental and Occupational Health, Indiana School of Public Health, Indiana University, Bloomington, Indiana, USA
| | - Arun R Pandiri
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | | | - Julia E Rager
- The Institute for Environmental Health Solutions, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Keith Q Tanis
- Safety Assessment and Laboratory Animal Resources, Merck Sharp & Dohme Corp, West Point, Pennsylvania, USA
| | - Jan Willem van der Laan
- Section on Pharmacology, Toxicology and Kinetics, Medicines Evaluation Board, Utrecht, The Netherlands
| | - Alisa Vespa
- Therapeutic Products Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Carole L Yauk
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Syril D Pettit
- Health and Environmental Sciences Institute, Washington, District of Columbia, USA
| | - Frank D Sistare
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
30
|
Satake K, Ishii T, Morikawa T, Sakamoto T, Nishii Y. Quercetin Reduces the Development of 2,3,7,8-Tetrachlorodibenzo-p-dioxin-Induced Cleft Palate in Mice by Suppressing CYP1A1 via the Aryl Hydrocarbon Receptor. Nutrients 2022; 14:nu14122448. [PMID: 35745180 PMCID: PMC9229746 DOI: 10.3390/nu14122448] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 06/07/2022] [Accepted: 06/10/2022] [Indexed: 11/16/2022] Open
Abstract
Quercetin is a flavonoid with a wide range of pharmacological activities, including anticancer, antioxidant, and anti-inflammatory effects. Since it is a nutrient that can be consumed with a regular diet, quercetin has recently garnered interest. Quercetin acts as a phytochemical ligand for the aryl hydrocarbon receptor (AhR). Cleft lip and palate are among the most frequently diagnosed congenital diseases, and exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) during pregnancy induces cleft palate via AhR. In this study, we investigated the preventive effect of quercetin intake on the TCDD-induced cleft palate and its mechanism of action. The in vivo results suggest that quercetin intake by pregnant mice can prevent cleft palate in fetal mice. In vitro, the addition of TCDD induced a reduction in cell migration and the proliferation of mouse embryonic palatal mesenchymal cells, which was mitigated by the addition of quercetin. The addition of quercetin did not alter the mRNA expression levels of the AhR repressor but significantly suppressed mRNA expression of CYP1A1. In addition, the binding of AhR to a xenobiotic responsive element was inhibited by quercetin, based on a chemically activated luciferase expression assay. In conclusion, our results suggest that quercetin reduces the development of TCDD-induced cleft palate by inhibiting CYP1A1 through AhR.
Collapse
|
31
|
Current Therapeutic Landscape and Safety Roadmap for Targeting the Aryl Hydrocarbon Receptor in Inflammatory Gastrointestinal Indications. Cells 2022; 11:cells11101708. [PMID: 35626744 PMCID: PMC9139855 DOI: 10.3390/cells11101708] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/30/2022] [Accepted: 05/16/2022] [Indexed: 02/07/2023] Open
Abstract
Target modulation of the AhR for inflammatory gastrointestinal (GI) conditions holds great promise but also the potential for safety liabilities both within and beyond the GI tract. The ubiquitous expression of the AhR across mammalian tissues coupled with its role in diverse signaling pathways makes development of a “clean” AhR therapeutically challenging. Ligand promiscuity and diversity in context-specific AhR activation further complicates targeting the AhR for drug development due to limitations surrounding clinical translatability. Despite these concerns, several approaches to target the AhR have been explored such as small molecules, microbials, PROTACs, and oligonucleotide-based approaches. These various chemical modalities are not without safety liabilities and require unique de-risking strategies to parse out toxicities. Collectively, these programs can benefit from in silico and in vitro methodologies that investigate specific AhR pathway activation and have the potential to implement thresholding parameters to categorize AhR ligands as “high” or “low” risk for sustained AhR activation. Exploration into transcriptomic signatures for AhR safety assessment, incorporation of physiologically-relevant in vitro model systems, and investigation into chronic activation of the AhR by structurally diverse ligands will help address gaps in our understanding regarding AhR-dependent toxicities. Here, we review the role of the AhR within the GI tract, novel therapeutic modality approaches to target the AhR, key AhR-dependent safety liabilities, and relevant strategies that can be implemented to address drug safety concerns. Together, this review discusses the emerging therapeutic landscape of modalities targeting the AhR for inflammatory GI indications and offers a safety roadmap for AhR drug development.
Collapse
|
32
|
Gupta V, Hammond CL, Roztocil E, Gonzalez MO, Feldon SE, Woeller CF. Thinking inside the box: Current insights into targeting orbital tissue remodeling and inflammation in thyroid eye disease. Surv Ophthalmol 2022; 67:858-874. [PMID: 34487739 PMCID: PMC8891393 DOI: 10.1016/j.survophthal.2021.08.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 08/24/2021] [Accepted: 08/30/2021] [Indexed: 12/21/2022]
Abstract
Thyroid eye disease (TED) is an autoimmune disorder that manifests in the orbit. In TED, the connective tissue behind the eye becomes inflamed and remodels with increased fat accumulation and/or increased muscle and scar tissue. As orbital tissue expands, patients develop edema, exophthalmos, diplopia, and optic neuropathy. In severe cases vision loss may occur secondary to corneal scarring from exposure or optic nerve compression. Currently there is no cure for TED, and treatments are limited. A major breakthrough in TED therapy occurred with the FDA approval of teprotumumab, a monoclonal insulin-like growth factor 1 receptor (IGF1R) blocking antibody. Yet, teprotumumab therapy has limitations, including cost, infusion method of drug delivery, variable response, and relapse. We describe approaches to target orbital fibroblasts and the complex pathophysiology that underlies tissue remodeling and inflammation driving TED. Further advances in the elucidation of the mechanisms of TED may lead to prophylaxis based upon early biomarkers as well as lead to more convenient, less expensive therapies.
Collapse
Affiliation(s)
- Vardaan Gupta
- Flaum Eye Institute, University of Rochester, 210 Crittenden Boulevard, Rochester, New York 14642, USA
| | - Christine L Hammond
- Flaum Eye Institute, University of Rochester, 210 Crittenden Boulevard, Rochester, New York 14642, USA
| | - Elisa Roztocil
- Flaum Eye Institute, University of Rochester, 210 Crittenden Boulevard, Rochester, New York 14642, USA
| | - Mithra O Gonzalez
- Flaum Eye Institute, University of Rochester, 210 Crittenden Boulevard, Rochester, New York 14642, USA
| | - Steven E Feldon
- Flaum Eye Institute, University of Rochester, 210 Crittenden Boulevard, Rochester, New York 14642, USA
| | - Collynn F Woeller
- Flaum Eye Institute, University of Rochester, 210 Crittenden Boulevard, Rochester, New York 14642, USA.
| |
Collapse
|
33
|
Zhang W, Xie HQ, Li Y, Zhou M, Zhou Z, Wang R, Hahn ME, Zhao B. The aryl hydrocarbon receptor: A predominant mediator for the toxicity of emerging dioxin-like compounds. JOURNAL OF HAZARDOUS MATERIALS 2022; 426:128084. [PMID: 34952507 PMCID: PMC9039345 DOI: 10.1016/j.jhazmat.2021.128084] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/29/2021] [Accepted: 12/12/2021] [Indexed: 06/01/2023]
Abstract
The aryl hydrocarbon receptor (AHR) is a member of the basic helix-loop-helix/Per-ARNT-Sim (bHLH-PAS) family of transcription factors and has broad biological functions. Early after the identification of the AHR, most studies focused on its roles in regulating the expression of drug-metabolizing enzymes and mediating the toxicity of dioxins and dioxin-like compounds (DLCs). Currently, more diverse functions of AHR have been identified, indicating that AHR is not just a dioxin receptor. Dioxins and DLCs occur ubiquitously and have diverse health/ecological risks. Additional research is required to identify both shared and compound-specific mechanisms, especially for emerging DLCs such as polyhalogenated carbazoles (PHCZs), polychlorinated diphenyl sulfides (PCDPSs), and others, of which only a few investigations have been performed at present. Many of the toxic effects of emerging DLCs were observed to be predominantly mediated by the AHR because of their structural similarity as dioxins, and the in vitro TCDD-relative potencies of certain emerging DLC congeners are comparable to or even greater than the WHO-TEFs of OctaCDD, OctaCDF, and most coplanar PCBs. Due to the close relationship between AHR biology and environmental science, this review begins by providing novel insights into AHR signaling (canonical and non-canonical), AHR's biochemical properties (AHR structure, AHR-ligand interaction, AHR-DNA binding), and the variations during AHR transactivation. Then, AHR ligand classification and the corresponding mechanisms are discussed, especially the shared and compound-specific, AHR-mediated effects and mechanisms of emerging DLCs. Accordingly, a series of in vivo and in vitro toxicity evaluation methods based on the AHR signaling pathway are reviewed. In light of current advances, future research on traditional and emerging DLCs will enhance our understanding of their mechanisms, toxicity, potency, and ecological impacts.
Collapse
Affiliation(s)
- Wanglong Zhang
- College of Life Sciences, Qufu Normal University, Qufu, Shandong 273165, 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
| | - Yunping Li
- 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
| | - Mingxi Zhou
- Biology Centre of the Czech Academy of Sciences v.v.i, Institute of Plant Molecular Biology, Branišovská 31, 370 05 České Budějovice, Czech Republic
| | - Zhiguang Zhou
- State Environmental Protection Key Laboratory of Dioxin Pollution Control, National Research Center for Environmental Analysis and Measurement, Beijing 100029, China
| | - Renjun Wang
- College of Life Sciences, Qufu Normal University, Qufu, Shandong 273165, China
| | - Mark E Hahn
- Biology Department, Woods Hole Oceanographic Institution (WHOI), Woods Hole, MA 02543, USA; Boston University Superfund Research Program, Boston University, Boston, MA 02118, USA
| | - 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.
| |
Collapse
|
34
|
Hammond CL, Roztocil E, Gupta V, Feldon SE, Woeller CF. More than Meets the Eye: The Aryl Hydrocarbon Receptor is an Environmental Sensor, Physiological Regulator and a Therapeutic Target in Ocular Disease. FRONTIERS IN TOXICOLOGY 2022; 4:791082. [PMID: 35295218 PMCID: PMC8915869 DOI: 10.3389/ftox.2022.791082] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/08/2022] [Indexed: 12/22/2022] Open
Abstract
The aryl hydrocarbon receptor (AHR) is a ligand activated transcription factor originally identified as an environmental sensor of xenobiotic chemicals. However, studies have revealed that the AHR regulates crucial aspects of cell growth and metabolism, development and the immune system. The importance of the AHR and AHR signaling in eye development, toxicology and disease is now being uncovered. The AHR is expressed in many ocular tissues including the retina, choroid, cornea and the orbit. A significant role for the AHR in age-related macular degeneration (AMD), autoimmune uveitis, and other ocular diseases has been identified. Ligands for the AHR are structurally diverse organic molecules from exogenous and endogenous sources. Natural AHR ligands include metabolites of tryptophan and byproducts of the microbiome. Xenobiotic AHR ligands include persistent environmental pollutants such as dioxins, benzo (a) pyrene [B (a) P] and polychlorinated biphenyls (PCBs). Pharmaceutical agents including the proton pump inhibitors, esomeprazole and lansoprazole, and the immunosuppressive drug, leflunomide, activate the AHR. In this review, we highlight the role of the AHR in the eye and discuss how AHR signaling is involved in responding to endogenous and environmental stimuli. We also present the emerging concept that the AHR is a promising therapeutic target for eye disease.
Collapse
Affiliation(s)
| | | | | | | | - Collynn F. Woeller
- Flaum Eye Institute, Rochester, NY, United States
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester, Rochester, NY, United States
- *Correspondence: Collynn F. Woeller,
| |
Collapse
|
35
|
Franciosi JP, Mougey EB, Dellon ES, Gutierrez-Junquera C, Fernandez-Fernandez S, Venkatesh RD, Gupta SK. Proton Pump Inhibitor Therapy for Eosinophilic Esophagitis: History, Mechanisms, Efficacy, and Future Directions. J Asthma Allergy 2022; 15:281-302. [PMID: 35250281 PMCID: PMC8892718 DOI: 10.2147/jaa.s274524] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 01/14/2022] [Indexed: 12/11/2022] Open
Abstract
Over the past decade, the role of proton pump inhibitor (PPI) medication has evolved from a diagnostic tool for Eosinophilic Esophagitis (EoE), by excluding patients with PPI responsive esophageal eosinophilia (PPI-REE), to a therapy for EoE. This transition resulted from the Updated International Consensus Diagnostic Criteria for Eosinophilic Esophagitis: Proceedings of the Appraisal of Guidelines for Research and Evaluation II (AGREE) Conference to support PPI therapy for EoE in children and adults. Additional recent advances have suggested a role for genetic variations that might impact response to PPI therapy for EoE. This review article will explore a brief background of EoE, the evolution of PPI therapy for EoE and its proposed mechanisms, efficacy and safety in children and adults, and considerations for future PPI precision medicine in patients with EoE.
Collapse
Affiliation(s)
- James P Franciosi
- Division of Gastroenterology, Nemours Children’s Hospital, Orlando, FL, USA
- College of Medicine, University of Central Florida, Orlando, FL, USA
- Correspondence: James P Franciosi, Division of Gastroenterology, Nemours Children’s Hospital, 6535 Nemours Parkway, Orlando, FL, 32827, USA, Email
| | - Edward B Mougey
- Center for Pharmacogenomics and Translational Research, Nemours Children’s Health System, Jacksonville, FL, USA
| | - Evan S Dellon
- Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Carolina Gutierrez-Junquera
- Pediatric Gastroenterology Unit, Hospital Universitario Puerta de Hierro-Majadahonda, Autonomous University of Madrid, Madrid, Spain
| | | | - Rajitha D Venkatesh
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Nationwide Children’s Hospital, Columbus, OH, USA
| | - Sandeep K Gupta
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Riley Hospital for Children, Indiana University School of Medicine and Community Health Network, Indianapolis, IN, USA
| |
Collapse
|
36
|
Coelho NR, Pimpão AB, Correia MJ, Rodrigues TC, Monteiro EC, Morello J, Pereira SA. Pharmacological blockage of the AHR-CYP1A1 axis: a call for in vivo evidence. J Mol Med (Berl) 2021; 100:215-243. [PMID: 34800164 PMCID: PMC8605459 DOI: 10.1007/s00109-021-02163-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 10/27/2021] [Accepted: 11/03/2021] [Indexed: 01/21/2023]
Abstract
The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that can be activated by structurally diverse compounds arising from the environment and the microbiota and host metabolism. Expanding evidence has been shown that the modulation of the canonical pathway of AHR occurs during several chronic diseases and that its abrogation might be of clinical interest for metabolic and inflammatory pathological processes. However, most of the evidence on the pharmacological abrogation of the AHR-CYP1A1 axis has been reported in vitro, and therefore, guidance for in vivo studies is needed. In this review, we cover the state-of-the-art of the pharmacodynamic and pharmacokinetic properties of AHR antagonists and CYP1A1 inhibitors in different in vivo rodent (mouse or rat) models of disease. This review will serve as a road map for those researchers embracing this emerging therapeutic area targeting the AHR. Moreover, it is a timely opportunity as the first AHR antagonists have recently entered the clinical stage of drug development.
Collapse
Affiliation(s)
- N R Coelho
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056, Lisboa, Portugal
| | - A B Pimpão
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056, Lisboa, Portugal
| | - M J Correia
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056, Lisboa, Portugal
| | - T C Rodrigues
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056, Lisboa, Portugal
| | - E C Monteiro
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056, Lisboa, Portugal
| | - J Morello
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056, Lisboa, Portugal
| | - S A Pereira
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056, Lisboa, Portugal.
| |
Collapse
|
37
|
Lim TX, Ahamed M, Reutens DC. The aryl hydrocarbon receptor: A diagnostic and therapeutic target in glioma. Drug Discov Today 2021; 27:422-435. [PMID: 34624509 DOI: 10.1016/j.drudis.2021.09.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 07/29/2021] [Accepted: 09/29/2021] [Indexed: 12/19/2022]
Abstract
Glioblastoma multiforme (GBM) is a deadly disease; 5-year survival rates have shown little improvement over the past 30 years. In vivo positron emission tomography (PET) imaging is an important method of identifying potential diagnostic and therapeutic molecular targets non-invasively. The aryl hydrocarbon receptor (AhR) is a transcription factor that regulates multiple genes involved in immune response modulation and tumorigenesis. The AhR is an attractive potential drug target and studies have shown that its activation by small molecules can modulate innate and adaptive immunity beneficially and prevent AhR-mediated tumour promotion in several cancer types. In this review, we provide an overview of the role of the AhR in glioma tumorigenesis and highlight its potential as an emerging biomarker for glioma therapies targeting the tumour immune response and PET diagnostics.
Collapse
Affiliation(s)
- Ting Xiang Lim
- ARC Centre for Innovation in Biomedical Imaging Technology, Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia
| | - Muneer Ahamed
- ARC Centre for Innovation in Biomedical Imaging Technology, Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia
| | - David C Reutens
- ARC Centre for Innovation in Biomedical Imaging Technology, Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
38
|
Stoddard EG, Nag S, Martin J, Tyrrell KJ, Gibbins T, Anderson KA, Shukla AK, Corley R, Wright AT, Smith JN. Exposure to an Environmental Mixture of Polycyclic Aromatic Hydrocarbons Induces Hepatic Cytochrome P450 Enzymes in Mice. Chem Res Toxicol 2021; 34:2145-2156. [PMID: 34472326 DOI: 10.1021/acs.chemrestox.1c00235] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cytochrome P450 enzymes (CYPs) play an important role in bioactivating or detoxifying polycyclic aromatic hydrocarbons (PAHs), common environmental contaminants. While it is widely accepted that exposure to PAHs induces CYPs, effectively increasing rates of xenobiotic metabolism, dose- and time-response patterns of CYP induction are not well-known. In order to better understand dose- and time-response relationships of individual CYPs following induction, we exposed B6129SF1/J mice to single or repeated doses (2-180 μmol/kg/d) of benzo[a]pyrene (BaP) or Supermix-10, a mixture of the top 10 most abundant PAHs found at the Portland Harbor Superfund Site. In hepatic microsomes from exposed mice, we measured amounts of active CYPs using activity-based protein profiling and total CYP expression using global proteomics. We observed rapid Cyp1a1 induction after 6 h at the lowest PAH exposures and broad induction of many CYPs after 3 daily PAH doses at 72 h following the first dose. Using samples displaying Cyp1a1 induction, we observed significantly higher metabolic affinity for BaP metabolism (Km reduced 3-fold), 3-fold higher intrinsic clearance, but no changes to the Vmax. Mice dosed with the highest PAH exposures exhibited 1.7-5-fold higher intrinsic clearance rates for BaP compared to controls and higher Vmax values indicating greater amounts of enzymes capable of metabolizing BaP. This study demonstrates exposure to PAHs found at superfund sites induces enzymes in dose- and time-dependent patterns in mice. Accounting for specific changes in enzyme profiles, relative rates of PAH bioactivation and detoxification, and resulting risk will help translate internal dosimetry of animal models to humans and improve risk assessments of PAHs at superfund sites.
Collapse
Affiliation(s)
- Ethan G Stoddard
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Subhasree Nag
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Jude Martin
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Kimberly J Tyrrell
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Teresa Gibbins
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Kim A Anderson
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon 97331, United States
| | - Anil K Shukla
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Richard Corley
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Aaron T Wright
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352, United States.,The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, Washington 99163, United States
| | - Jordan N Smith
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99352, United States.,Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon 97331, United States
| |
Collapse
|
39
|
Correia MJ, Pimpão AB, Lopes-Coelho F, Sequeira CO, Coelho NR, Gonçalves-Dias C, Barouki R, Coumoul X, Serpa J, Morello J, Monteiro EC, Pereira SA. Aryl Hydrocarbon Receptor and Cysteine Redox Dynamics Underlie (Mal)adaptive Mechanisms to Chronic Intermittent Hypoxia in Kidney Cortex. Antioxidants (Basel) 2021; 10:antiox10091484. [PMID: 34573115 PMCID: PMC8469308 DOI: 10.3390/antiox10091484] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 12/31/2022] Open
Abstract
We hypothesized that an interplay between aryl hydrocarbon receptor (AhR) and cysteine-related thiolome at the kidney cortex underlies the mechanisms of (mal)adaptation to chronic intermittent hypoxia (CIH), promoting arterial hypertension (HTN). Using a rat model of CIH-HTN, we investigated the impact of short-term (1 and 7 days), mid-term (14 and 21 days, pre-HTN), and long-term intermittent hypoxia (IH) (up to 60 days, established HTN) on CYP1A1 protein level (a sensitive hallmark of AhR activation) and cysteine-related thiol pools. We found that acute and chronic IH had opposite effects on CYP1A1 and the thiolome. While short-term IH decreased CYP1A1 and increased protein-S-thiolation, long-term IH increased CYP1A1 and free oxidized cysteine. In addition, an in vitro administration of cystine, but not cysteine, to human endothelial cells increased Cyp1a1 expression, supporting cystine as a putative AhR activator. This study supports CYP1A1 as a biomarker of obstructive sleep apnea (OSA) severity and oxidized pools of cysteine as risk indicator of OSA-HTN. This work contributes to a better understanding of the mechanisms underlying the phenotype of OSA-HTN, mimicked by this model, which is in line with precision medicine challenges in OSA.
Collapse
Affiliation(s)
- Maria João Correia
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (F.L.-C.); (C.O.S.); (N.R.C.); (C.G.-D.); (J.S.); (J.M.); (E.C.M.)
| | - António B. Pimpão
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (F.L.-C.); (C.O.S.); (N.R.C.); (C.G.-D.); (J.S.); (J.M.); (E.C.M.)
| | - Filipa Lopes-Coelho
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (F.L.-C.); (C.O.S.); (N.R.C.); (C.G.-D.); (J.S.); (J.M.); (E.C.M.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023 Lisboa, Portugal
| | - Catarina O. Sequeira
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (F.L.-C.); (C.O.S.); (N.R.C.); (C.G.-D.); (J.S.); (J.M.); (E.C.M.)
| | - Nuno R. Coelho
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (F.L.-C.); (C.O.S.); (N.R.C.); (C.G.-D.); (J.S.); (J.M.); (E.C.M.)
| | - Clara Gonçalves-Dias
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (F.L.-C.); (C.O.S.); (N.R.C.); (C.G.-D.); (J.S.); (J.M.); (E.C.M.)
| | - Robert Barouki
- INSERM UMR-S 1124, 3TS, Environmental Toxicity, Therapeutic Targets, Cellular Signaling and Biomarkers, Université de Paris, 45 rue des Saints-Pères, 75006 Paris, France; (R.B.); (X.C.)
| | - Xavier Coumoul
- INSERM UMR-S 1124, 3TS, Environmental Toxicity, Therapeutic Targets, Cellular Signaling and Biomarkers, Université de Paris, 45 rue des Saints-Pères, 75006 Paris, France; (R.B.); (X.C.)
| | - Jacinta Serpa
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (F.L.-C.); (C.O.S.); (N.R.C.); (C.G.-D.); (J.S.); (J.M.); (E.C.M.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023 Lisboa, Portugal
| | - Judit Morello
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (F.L.-C.); (C.O.S.); (N.R.C.); (C.G.-D.); (J.S.); (J.M.); (E.C.M.)
| | - Emília C. Monteiro
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (F.L.-C.); (C.O.S.); (N.R.C.); (C.G.-D.); (J.S.); (J.M.); (E.C.M.)
| | - Sofia A. Pereira
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (F.L.-C.); (C.O.S.); (N.R.C.); (C.G.-D.); (J.S.); (J.M.); (E.C.M.)
- Correspondence:
| |
Collapse
|
40
|
Stockinger B, Shah K, Wincent E. AHR in the intestinal microenvironment: safeguarding barrier function. Nat Rev Gastroenterol Hepatol 2021; 18:559-570. [PMID: 33742166 PMCID: PMC7611426 DOI: 10.1038/s41575-021-00430-8] [Citation(s) in RCA: 205] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/09/2021] [Indexed: 02/01/2023]
Abstract
Mammalian aryl hydrocarbon receptor (AHR) is a ligand-dependent transcription factor that belongs to the basic helix-loop-helix (bHLH)-PAS family of transcription factors, which are evolutionarily conserved environmental sensors. In the absence of ligands, AHR resides in the cytoplasm in a complex with molecular chaperones such as HSP90, XAP2 and p23. Upon ligand binding, AHR translocates into the nuclear compartment, where it dimerizes with its partner protein, AHR nuclear translocator (ARNT), an obligatory partner for the DNA-binding and functional activity. Historically, AHR had mostly been considered as a key intermediary for the detrimental effects of environmental pollutants on the body. However, following the discovery of AHR-mediated functions in various immune cells, as well as the emergence of non-toxic 'natural' AHR ligands, this view slowly began to change, and the study of AHR-deficient mice revealed a plethora of important beneficial functions linked to AHR activation. This Review focuses on regulation of the AHR pathway and the barrier-protective roles AHR has in haematopoietic, as well as non-haematopoietic, cells within the intestinal microenvironment. It covers the nature of AHR ligands and feedback regulation of the AHR pathway, outlining the currently known physiological functions in immune, epithelial, endothelial and neuronal cells of the intestine.
Collapse
Affiliation(s)
| | | | - Emma Wincent
- Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
41
|
Podtelezhnikov AA, Monroe JJ, Aslamkhan AG, Pearson K, Qin C, Tamburino AM, Loboda AP, Glaab WE, Sistare FD, Tanis KQ. Quantitative Transcriptional Biomarkers of Xenobiotic Receptor Activation in Rat Liver for the Early Assessment of Drug Safety Liabilities. Toxicol Sci 2021; 175:98-112. [PMID: 32119089 DOI: 10.1093/toxsci/kfaa026] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The robust transcriptional plasticity of liver mediated through xenobiotic receptors underlies its ability to respond rapidly and effectively to diverse chemical stressors. Thus, drug-induced gene expression changes in liver serve not only as biomarkers of liver injury, but also as mechanistic sentinels of adaptation in metabolism, detoxification, and tissue protection from chemicals. Modern RNA sequencing methods offer an unmatched opportunity to quantitatively monitor these processes in parallel and to contextualize the spectrum of dose-dependent stress, adaptation, protection, and injury responses induced in liver by drug treatments. Using this approach, we profiled the transcriptional changes in rat liver following daily oral administration of 120 different compounds, many of which are known to be associated with clinical risk for drug-induced liver injury by diverse mechanisms. Clustering, correlation, and linear modeling analyses were used to identify and optimize coexpressed gene signatures modulated by drug treatment. Here, we specifically focused on prioritizing 9 key signatures for their pragmatic utility for routine monitoring in initial rat tolerability studies just prior to entering drug development. These signatures are associated with 5 canonical xenobiotic nuclear receptors (AHR, CAR, PXR, PPARα, ER), 3 mediators of reactive metabolite-mediated stress responses (NRF2, NRF1, P53), and 1 liver response following activation of the innate immune response. Comparing paradigm chemical inducers of each receptor to the other compounds surveyed enabled us to identify sets of optimized gene expression panels and associated scoring algorithms proposed as quantitative mechanistic biomarkers with high sensitivity, specificity, and quantitative accuracy. These findings were further qualified using public datasets, Open TG-GATEs and DrugMatrix, and internal development compounds. With broader collaboration and additional qualification, the quantitative toxicogenomic framework described here could inform candidate selection prior to committing to drug development, as well as complement and provide a deeper understanding of the conventional toxicology study endpoints used later in drug development.
Collapse
Affiliation(s)
| | - James J Monroe
- Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania 19486-0004
| | - Amy G Aslamkhan
- Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania 19486-0004
| | - Kara Pearson
- Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania 19486-0004
| | - Chunhua Qin
- Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania 19486-0004
| | | | | | - Warren E Glaab
- Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania 19486-0004
| | - Frank D Sistare
- Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania 19486-0004
| | | |
Collapse
|
42
|
O'Donnell EF, Jang HS, Liefwalker DF, Kerkvliet NI, Kolluri SK. Discovery and Mechanistic Characterization of a Select Modulator of AhR-regulated Transcription (SMAhRT) with Anti-cancer Effects. Apoptosis 2021; 26:307-322. [PMID: 33893898 DOI: 10.1007/s10495-021-01666-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2021] [Indexed: 02/07/2023]
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor and a member of the bHLH/PAS (basic Helix-Loop-Helix/Per-Arnt-Sim) family of proteins. The AhR was cloned and characterized for its role in mediating the toxicity of dioxins. Subsequent research has identified the role of AhR in suppression of cancer cell growth. We hypothesized that the AhR is a molecular target for therapeutic intervention in cancer, and that activation of the AhR by unique AhR ligands in cancer cells could have anti-cancer effects including induction of cell death. This study describes the discovery and characterization of a new class of anti-cancer agents targeting the AhR, that we designate as Select Modulators of AhR-regulated Transcription (SMAhRTs). We employed two independent small molecule screening approaches to identify potential SMAhRTs. We report the identification of CGS-15943 that activates AhR signaling and induces apoptosis in an AhR-dependent manner in liver and breast cancer cells. Investigation of the downstream signaling pathway of this newly identified SMAhRT revealed upregulation of Fas-ligand (FasL), which is required for AhR-mediated apoptosis. Our results provide a basis for further development of a new class of anti-cancer therapeutics targeting an underappreciated molecular target, the AhR.
Collapse
Affiliation(s)
- Edmond Francis O'Donnell
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, 97331, USA
- Department of Orthopaedic Surgery, University of California Davis Medical Center, Davis, CA, USA
| | - Hyo Sang Jang
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, 97331, USA
| | - Daniel F Liefwalker
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, 97331, USA
- Department of Molecular and Medical Genetics, School of Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Nancy I Kerkvliet
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, 97331, USA
- The Pacific Northwest Center for Translational Environmental Health Research, Oregon State University, Corvallis, OR, 97331, USA
| | - Siva Kumar 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.
- Center for Genome Research and Biocomputing, Oregon State University, Corvallis, OR, 97331, USA.
- The Pacific Northwest Center for Translational Environmental Health Research, Oregon State University, Corvallis, OR, 97331, USA.
| |
Collapse
|
43
|
Blevins LK, Zhou J, Crawford RB, Kaminski NE. Identification of a Sensitive Human Immunological Target of Aryl Hydrocarbon Receptor Activation: CD5 + Innate-Like B Cells. Front Immunol 2021; 12:635748. [PMID: 33936048 PMCID: PMC8082145 DOI: 10.3389/fimmu.2021.635748] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/24/2021] [Indexed: 01/02/2023] Open
Abstract
Xenobiotic-mediated activation of the aryl hydrocarbon receptor (AHR) is immunotoxic in a number of immune cell types, with the B cell being a well-established sensitive target. Recent advances have provided evidence that the B cell repertoire is a heterogeneous population, with subpopulations exhibiting vastly different cellular and functional phenotypes. Recent work from our laboratory identified the T cell specific kinase lck as being differentially regulated by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), which is a potent activator of AHR. While LCK is primarily expressed in T cells, a subset of CD5+ B cells also express LCK. CD5 positivity describes a broad class of B lymphocytes termed innate-like B cells (ILBs) that are critical mediators of innate immunity through constitutive secretion of polyvalent natural immunoglobulin M (IgM). We hypothesized that CD5+ ILBs may be sensitive to AHR-mediated immunotoxicity. Indeed, when CD5+ B cells were isolated from the CD19+ pool and treated with TCDD, they showed increased suppression of the CD40 ligand-induced IgM response compared to CD5- B cells. Further, characterization of the CD5+ population indicated increased basal expression of AHR, AHR repressor (AHRR), and cytochrome p450 family 1 member a1 (CYP1A1). Indeed the levels of AHR-mediated suppression of the IgM response from individual donors strongly correlated with the percentage of the B cell pool that was CD5+, suggesting that CD5+ B cells are more sensitive to AHR-mediated impairment. Together these data highlight the sensitive nature of CD5+ ILBs to AHR activation and provide insight into mechanisms associated with AHR activation in human B cells.
Collapse
Affiliation(s)
- Lance K Blevins
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
| | - Jiajun Zhou
- Department of Microbiology & Molecular Genetics, Michigan State University, East Lansing, MI, United States
| | - Robert B Crawford
- Department of Toxicology & Pharmacology, Michigan State University, East Lansing, MI, United States
| | - Norbert E Kaminski
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States.,Department of Toxicology & Pharmacology, Michigan State University, East Lansing, MI, United States.,Center for Research on Ingredient Safety, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
44
|
Esteban J, Sánchez-Pérez I, Hamscher G, Miettinen HM, Korkalainen M, Viluksela M, Pohjanvirta R, Håkansson H. Role of aryl hydrocarbon receptor (AHR) in overall retinoid metabolism: Response comparisons to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure between wild-type and AHR knockout mice. Reprod Toxicol 2021; 101:33-49. [PMID: 33607186 DOI: 10.1016/j.reprotox.2021.02.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 01/20/2021] [Accepted: 02/10/2021] [Indexed: 02/06/2023]
Abstract
Young adult wild-type and aryl hydrocarbon receptor knockout (AHRKO) mice of both sexes and the C57BL/6J background were exposed to 10 weekly oral doses of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD; total dose of 200 μg/kg bw) to further characterize the observed impacts of AHR as well as TCDD on the retinoid system. Unexposed AHRKO mice harboured heavier kidneys, lighter livers and lower serum all-trans retinoic acid (ATRA) and retinol (REOH) concentrations than wild-type mice. Results from the present study also point to a role for the murine AHR in the control of circulating REOH and ATRA concentrations. In wild-type mice, TCDD elevated liver weight and reduced thymus weight, and drastically reduced the hepatic concentrations of 9-cis-4-oxo-13,14-dihydro-retinoic acid (CORA) and retinyl palmitate (REPA). In female wild-type mice, TCDD increased the hepatic concentration of ATRA as well as the renal and circulating REOH concentrations. Renal CORA concentrations were substantially diminished in wild-type male mice exclusively following TCDD-exposure, with a similar tendency in serum. In contrast, TCDD did not affect any of these toxicity or retinoid system parameters in AHRKO mice. Finally, a distinct sex difference occurred in kidney concentrations of all the analysed retinoid forms. Together, these results strengthen the evidence of a mandatory role of AHR in TCDD-induced retinoid disruption, and suggest that the previously reported accumulation of several retinoid forms in the liver of AHRKO mice is a line-specific phenomenon. Our data further support participation of AHR in the control of liver and kidney development in mice.
Collapse
Affiliation(s)
- Javier Esteban
- Instituto De Bioingeniería, Universidad Miguel Hernández De Elche, Elche, Alicante, Spain.
| | - Ismael Sánchez-Pérez
- Instituto De Bioingeniería, Universidad Miguel Hernández De Elche, Elche, Alicante, Spain.
| | - Gerd Hamscher
- Institute of Food Chemistry and Food Biotechnology, Justus Liebig University Giessen, Giessen, Germany.
| | - Hanna M Miettinen
- School of Pharmacy (Toxicology) and Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland.
| | - Merja Korkalainen
- Environmental Health Unit, Finnish Insitute for Health and Welfare (THL), Kuopio, Finland.
| | - Matti Viluksela
- School of Pharmacy (Toxicology) and Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland; Environmental Health Unit, Finnish Insitute for Health and Welfare (THL), Kuopio, Finland.
| | - Raimo Pohjanvirta
- Department of Food Hygiene & Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, Mustialankatu 1, FI-00790 Helsinki, Finland.
| | - Helen Håkansson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
45
|
Goedtke L, Sprenger H, Hofmann U, Schmidt FF, Hammer HS, Zanger UM, Poetz O, Seidel A, Braeuning A, Hessel-Pras S. Polycyclic Aromatic Hydrocarbons Activate the Aryl Hydrocarbon Receptor and the Constitutive Androstane Receptor to Regulate Xenobiotic Metabolism in Human Liver Cells. Int J Mol Sci 2020; 22:ijms22010372. [PMID: 33396476 PMCID: PMC7796163 DOI: 10.3390/ijms22010372] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/18/2020] [Accepted: 12/26/2020] [Indexed: 12/19/2022] Open
Abstract
Polycyclic aromatic hydrocarbons (PAHs) are environmental pollutants produced by incomplete combustion of organic matter. They induce their own metabolism by upregulating xenobiotic-metabolizing enzymes such as cytochrome P450 monooxygenase 1A1 (CYP1A1) by activating the aryl hydrocarbon receptor (AHR). However, previous studies showed that individual PAHs may also interact with the constitutive androstane receptor (CAR). Here, we studied ten PAHs, different in carcinogenicity classification, for their potential to activate AHR- and CAR-dependent luciferase reporter genes in human liver cells. The majority of investigated PAHs activated AHR, while non-carcinogenic PAHs tended to activate CAR. We further characterized gene expression, protein abundancies and activities of the AHR targets CYP1A1 and 1A2, and the CAR target CYP2B6 in human HepaRG hepatoma cells. Enzyme induction patterns strongly resembled the profiles obtained at the receptor level, with AHR-activating PAHs inducing CYP1A1/1A2 and CAR-activating PAHs inducing CYP2B6. In summary, this study provides evidence that beside well-known activation of AHR, some PAHs also activate CAR, followed by subsequent expression of respective target genes. Furthermore, we found that an increased PAH ring number is associated with AHR activation as well as the induction of DNA double-strand breaks, whereas smaller PAHs activated CAR but showed no DNA-damaging potential.
Collapse
Affiliation(s)
- Lisa Goedtke
- Department Food Safety, German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Straße 8-10, 10589 Berlin, Germany; (L.G.); (H.S.); (A.B.)
| | - Heike Sprenger
- Department Food Safety, German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Straße 8-10, 10589 Berlin, Germany; (L.G.); (H.S.); (A.B.)
| | - Ute Hofmann
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Auerbachstr. 112, 70376 Stuttgart, and University of Tübingen, 72074 Tübingen, Germany; (U.H.); (U.M.Z.)
| | - Felix F. Schmidt
- SIGNATOPE GmbH, Markwiesenstraße 55, 72770 Reutlingen, Germany; (F.F.S.); (H.S.H.); (O.P.)
| | - Helen S. Hammer
- SIGNATOPE GmbH, Markwiesenstraße 55, 72770 Reutlingen, Germany; (F.F.S.); (H.S.H.); (O.P.)
| | - Ulrich M. Zanger
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Auerbachstr. 112, 70376 Stuttgart, and University of Tübingen, 72074 Tübingen, Germany; (U.H.); (U.M.Z.)
| | - Oliver Poetz
- SIGNATOPE GmbH, Markwiesenstraße 55, 72770 Reutlingen, Germany; (F.F.S.); (H.S.H.); (O.P.)
| | - Albrecht Seidel
- Biochemical Institute for Environmental Carcinogens, Prof. Dr. Gernot Grimmer Foundation, Lurup 4, 22927 Grosshansdorf, Germany;
| | - Albert Braeuning
- Department Food Safety, German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Straße 8-10, 10589 Berlin, Germany; (L.G.); (H.S.); (A.B.)
| | - Stefanie Hessel-Pras
- Department Food Safety, German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Straße 8-10, 10589 Berlin, Germany; (L.G.); (H.S.); (A.B.)
- Correspondence: ; Tel.: +49-30-18412-25203
| |
Collapse
|
46
|
Safe S, Jin UH, Park H, Chapkin RS, Jayaraman A. Aryl Hydrocarbon Receptor (AHR) Ligands as Selective AHR Modulators (SAhRMs). Int J Mol Sci 2020; 21:6654. [PMID: 32932962 PMCID: PMC7555580 DOI: 10.3390/ijms21186654] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/01/2020] [Accepted: 09/09/2020] [Indexed: 02/06/2023] Open
Abstract
The aryl hydrocarbon receptor (AhR) was first identified as the intracellular protein that bound and mediated the toxic effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD, dioxin) and dioxin-like compounds (DLCs). Subsequent studies show that the AhR plays an important role in maintaining cellular homeostasis and in pathophysiology, and there is increasing evidence that the AhR is an important drug target. The AhR binds structurally diverse compounds, including pharmaceuticals, phytochemicals and endogenous biochemicals, some of which may serve as endogenous ligands. Classification of DLCs and non-DLCs based on their persistence (metabolism), toxicities, binding to wild-type/mutant AhR and structural similarities have been reported. This review provides data suggesting that ligands for the AhR are selective AhR modulators (SAhRMs) that exhibit tissue/cell-specific AhR agonist and antagonist activities, and that their functional diversity is similar to selective receptor modulators that target steroid hormone and other nuclear receptors.
Collapse
Affiliation(s)
- Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA; (U.-h.J.); (H.P.)
| | - Un-ho Jin
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA; (U.-h.J.); (H.P.)
| | - Hyejin Park
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA; (U.-h.J.); (H.P.)
| | - Robert S. Chapkin
- Departments of Nutrition and Food Science and Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA;
| | - Arul Jayaraman
- Department of Chemical Engineering, Texas A&M University, College Station, TX 77843, USA;
| |
Collapse
|
47
|
Wajda A, Łapczuk-Romańska J, Paradowska-Gorycka A. Epigenetic Regulations of AhR in the Aspect of Immunomodulation. Int J Mol Sci 2020; 21:E6404. [PMID: 32899152 PMCID: PMC7504141 DOI: 10.3390/ijms21176404] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/27/2020] [Accepted: 08/28/2020] [Indexed: 02/07/2023] Open
Abstract
Environmental factors contribute to autoimmune disease manifestation, and as regarded today, AhR has become an important factor in studies of immunomodulation. Besides immunological aspects, AhR also plays a role in pharmacological, toxicological and many other physiological processes such as adaptive metabolism. In recent years, epigenetic mechanisms have provided new insight into gene regulation and reveal a new contribution to autoimmune disease pathogenesis. DNA methylation, histone modifications, chromatin alterations, microRNA and consequently non-genetic changes in phenotypes connect with environmental factors. Increasing data reveals AhR cross-roads with the most significant in immunology pathways. Although study on epigenetic modulations in autoimmune diseases is still not well understood, therefore future research will help us understand their pathophysiology and help to find new therapeutic strategies. Present literature review sheds the light on the common ground between remodeling chromatin compounds and autoimmune antibodies used in diagnostics. In the proposed review we summarize recent findings that describe epigenetic factors which regulate AhR activity and impact diverse immunological responses and pathological changes.
Collapse
Affiliation(s)
- Anna Wajda
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland;
| | - Joanna Łapczuk-Romańska
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, 70-111 Szczecin, Poland;
| | - Agnieszka Paradowska-Gorycka
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland;
| |
Collapse
|
48
|
First evidence of aryl hydrocarbon receptor as a druggable target in hypertension induced by chronic intermittent hypoxia. Pharmacol Res 2020; 159:104869. [DOI: 10.1016/j.phrs.2020.104869] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/15/2020] [Accepted: 04/24/2020] [Indexed: 02/08/2023]
|
49
|
Jin UH, Michelhaugh SK, Polin LA, Shrestha R, Mittal S, Safe S. Omeprazole Inhibits Glioblastoma Cell Invasion and Tumor Growth. Cancers (Basel) 2020; 12:2097. [PMID: 32731514 PMCID: PMC7465678 DOI: 10.3390/cancers12082097] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 02/06/2023] Open
Abstract
Background: The aryl hydrocarbon receptor (AhR) is expressed in gliomas and the highest staining is observed in glioblastomas. A recent study showed that the AhR exhibited tumor suppressor-like activity in established and patient-derived glioblastoma cells and genomic analysis showed that this was due, in part, to suppression of CXCL12, CXCR4 and MMP9. Methods: Selective AhR modulators (SAhRMs) including AhR-active pharmaceuticals were screened for their inhibition of invasion using a spheroid invasion assay in patient-derived AhR-expressing 15-037 glioblastoma cells and in AhR-silenced 15-037 cells. Invasion, migration and cell proliferation were determined using spheroid invasion, Boyden chambers and scratch assay, and XTT metabolic assays for cell growth. Changes in gene and gene product expression were determined by real-time PCR and Western blot assays, respectively. In vivo antitumorigenic activity of omeprazole was determined in SCID mice bearing subcutaneous patient-derived 15-037 cells. Results: Results of a screening assay using patient-derived 15-037 cells (wild-type and AhR knockout) identified the AhR-active proton pump inhibitor omeprazole as an inhibitor of glioblastoma cell invasion and migration only AhR-expressing cells but not in cells where the AhR was downregulated. Omeprazole also enhanced AhR-dependent repression of the pro-invasion CXCL12, CXCR4 and MMP9 genes, and interactions and effectiveness of omeprazole plus temozolomide were response-dependent. Omeprazole (100 mg/kg/injection) inhibited and delayed tumors in SCID mice bearing patient-derived 15-037 cells injected subcutaneously. Conclusion: Our results demonstrate that omeprazole enhances AhR-dependent inhibition of glioblastoma invasion and highlights a potential new avenue for development of a novel therapeutic mechanism-based approach for treating glioblastoma.
Collapse
Affiliation(s)
- Un-Ho Jin
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, USA;
| | - Sharon K. Michelhaugh
- Fralin Biomedical Research Institute, Virginia Tech Carilion School of Medicine, Roanoke, VA 24014, USA; (S.K.M.); (S.M.)
| | - Lisa A. Polin
- Department of Oncology, Wayne State University and Karmanos Cancer Institute, Detroit, MI 48201, USA;
| | - Rupesh Shrestha
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA;
| | - Sandeep Mittal
- Fralin Biomedical Research Institute, Virginia Tech Carilion School of Medicine, Roanoke, VA 24014, USA; (S.K.M.); (S.M.)
- Carilion Clinic-Neurosurgery, Roanoke, VA 24014, USA
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, USA;
| |
Collapse
|
50
|
Alternaria alternata Toxins Synergistically Activate the Aryl Hydrocarbon Receptor Pathway In Vitro. Biomolecules 2020; 10:biom10071018. [PMID: 32659980 PMCID: PMC7407958 DOI: 10.3390/biom10071018] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 06/28/2020] [Accepted: 06/30/2020] [Indexed: 02/06/2023] Open
Abstract
Alternaria molds simultaneously produce a large variety of mycotoxins, of which several were previously reported to induce enzymes of phase I metabolism through aryl hydrocarbon receptor activation. Thus, we investigated the potential of naturally occurring Alternaria toxin mixtures to induce Cytochrome P450 (CYP) 1A1/1A2/1B1 activity. Two variants of an extract from cultured Alternaria alternata, as well as the toxins alternariol (AOH), alternariol monomethyl ether (AME), altertoxin I (ATX-I), and altertoxin II (ATX-II), were tested singularly and in binary mixtures applying the 7-ethoxy-resorufin-O-deethylase (EROD) assay in MCF-7 breast cancer cells. Sub-cytotoxic concentrations of the two toxin mixtures, as well as ATX-I, ATX-II and AOH, exhibited dose-dependent enhancements of CYP 1 activity. ATX-I and ATX-II interacted synergistically in this respect, demonstrating the two perylene quinones as major contributors to the extract’s potential. Binary mixtures between AOH and the two altertoxins respectively exhibited concentration-dependent antagonistic as well as synergistic combinatory effects. Notably, AME showed no efficacy towards EROD enzyme activity or impact on other toxins’ efficacy. Hence, this study provides insights into synergistic and other combinatory effects of Alternaria toxins in natural co-occurrence scenarios in the context of AhR signalling pathway activation in breast cancer cells.
Collapse
|