1
|
Brewer G, Savage P, Fortier AM, Zhao H, Pacis A, Wang YC, Zuo D, de Nobrega M, Pedersen A, Cassel de Camps C, Souleimanova M, Ramos VM, Ragoussis J, Park M, Moraes C. Invasive phenotypes of triple-negative breast cancer-associated fibroblasts are mechanosensitive, AhR-dependent and correlate with disease state. Acta Biomater 2025:S1742-7061(25)00314-9. [PMID: 40318744 DOI: 10.1016/j.actbio.2025.04.061] [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: 11/26/2024] [Revised: 04/28/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
Cancer associated fibroblasts (CAFs) play a critically important role in facilitating tumour cell invasion during metastasis. They also modulate local biophysical features of the tumour microenvironment through the formation of fibrotic foci, which have been correlated with breast cancer aggression. However, the impact of the evolving three-dimensional biophysical tumour microenvironment on CAF function remains undefined. Here, by isolating CAFs from primary human triple-negative breast cancer tissue at the time of surgery, we find that their ability to remodel the local microenvironment and invade into a three-dimensional matrix correlates with disease state. We then engineered culture models to systematically deconstruct and recreate mechanical tissue features of early breast cancer fibrotic foci; and demonstrate that invasion is mechanically-activated only in CAFs from patients with no detectable pre-existing metastases, but is independent of mechanical cues in CAFs isolated from patients with later-stage axillary lymph node metastases. By comparing the differential transcriptional response of these cells to microenvironmental tissue stiffness, we identify the aryl hydrocarbon receptor (AhR) as being significantly upregulated in invasive sub-populations of both mechanically-activated and mechanically-insensitive CAFs. Increasing AhR expression in CAFs induced invasion, while suppressing AhR significantly reduced invasion in both mechanically-activated and mechanically-insensitive CAF populations, even on stiffnesses that recapitulate late-stage disease. This work therefore uses mechanobiological analyses to identify AhR as a mediator of CAF invasion, providing a potential stratification marker to identify those patients who might respond to future mechanics-based prophylactic therapies, and provides a targetable mechanism to limit CAF-associated metastatic disease progression in triple-negative breast cancer patients. STATEMENT OF SIGNIFICANCE: By designing a mechanically-tunable tissue-engineered model of fibroblastic foci, and using this to culture patient-derived cancer-associated fibroblasts, we demonstrate that these cells are differentially mechanosensitive, depending on disease stage of the patient. While comparing transcriptomic profiles of patient-derived cells produces too many pathways to screen, identifying the pathways activated by local tissue mechanics that were common across each patient allowed us to identify a specific target to limit fibroblast invasion. This broad discovery strategy may be useful across a variety of biomaterials-based tissue engineered models; and these specific findings suggest (1) a strategy to identify patients who might respond to CAF- or matrix-targeting therapies, and (2) a specific actionable target to limit CAF-associated metastatic disease progression.
Collapse
Affiliation(s)
- Gabrielle Brewer
- Rosalind & Morris Goodman Cancer Institute, McGill University, 1160 Avenues des Pins, Montréal, QC, Canada; Department of Biochemistry, McGill University, 3649 Promenade Sir-William-Osler, Montréal, QC, Canada
| | - Paul Savage
- Rosalind & Morris Goodman Cancer Institute, McGill University, 1160 Avenues des Pins, Montréal, QC, Canada; Department of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montréal, QC, Canada
| | - Anne-Marie Fortier
- Rosalind & Morris Goodman Cancer Institute, McGill University, 1160 Avenues des Pins, Montréal, QC, Canada
| | - Hong Zhao
- Rosalind & Morris Goodman Cancer Institute, McGill University, 1160 Avenues des Pins, Montréal, QC, Canada
| | - Alain Pacis
- Rosalind & Morris Goodman Cancer Institute, McGill University, 1160 Avenues des Pins, Montréal, QC, Canada
| | - Yu-Chang Wang
- Department of Human Genetics, McGill University, 3640 University, Montreal, QC; Genome Innovation Centre, 740 Dr Penfield Ave, Montreal, QC
| | - Dongmei Zuo
- Rosalind & Morris Goodman Cancer Institute, McGill University, 1160 Avenues des Pins, Montréal, QC, Canada
| | - Monyse de Nobrega
- Rosalind & Morris Goodman Cancer Institute, McGill University, 1160 Avenues des Pins, Montréal, QC, Canada; Department of Genetics and Molecular Biology, State University of Londrina, Londrina, PR, Brazil
| | - Annika Pedersen
- Rosalind & Morris Goodman Cancer Institute, McGill University, 1160 Avenues des Pins, Montréal, QC, Canada; Department of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montréal, QC, Canada
| | - Camille Cassel de Camps
- Department of Biomedical Engineering, McGill University, 3775 rue University, Montréal, QC, Canada
| | - Margarita Souleimanova
- Rosalind & Morris Goodman Cancer Institute, McGill University, 1160 Avenues des Pins, Montréal, QC, Canada
| | - Valentina Muñoz Ramos
- Rosalind & Morris Goodman Cancer Institute, McGill University, 1160 Avenues des Pins, Montréal, QC, Canada
| | - Jiannis Ragoussis
- Department of Human Genetics, McGill University, 3640 University, Montreal, QC; Genome Innovation Centre, 740 Dr Penfield Ave, Montreal, QC
| | - Morag Park
- Rosalind & Morris Goodman Cancer Institute, McGill University, 1160 Avenues des Pins, Montréal, QC, Canada; Department of Biochemistry, McGill University, 3649 Promenade Sir-William-Osler, Montréal, QC, Canada; Department of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montréal, QC, Canada; Department of Oncology, McGill University, 5100 de Maisonneuve Blvd. West, Montréal, QC, Canada.
| | - Christopher Moraes
- Rosalind & Morris Goodman Cancer Institute, McGill University, 1160 Avenues des Pins, Montréal, QC, Canada; Department of Biomedical Engineering, McGill University, 3775 rue University, Montréal, QC, Canada; Department of Chemical Engineering, McGill University, 3610 rue University, Montréal, QC, Canada.
| |
Collapse
|
2
|
Sulentic CEW, Kaplan BLF, Lawrence BP. Using the Key Characteristics Framework to Unlock the Mysteries of Aryl Hydrocarbon Receptor-Mediated Effects on the Immune System. Annu Rev Immunol 2025; 43:191-218. [PMID: 39813730 DOI: 10.1146/annurev-immunol-083122-040107] [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] [Indexed: 01/18/2025]
Abstract
Initially discovered for its role mediating the deleterious effects of environmental contaminants, the aryl hydrocarbon receptor (AHR) is now known to be a crucial regulator of the immune system. The expanding list of AHR ligands includes synthetic and naturally derived molecules spanning pollutants, phytochemicals, pharmaceuticals, and substances derived from amino acids and microorganisms. The consequences of engaging AHR vary, depending on factors such as the AHR ligand, cell type, immune challenge, developmental state, dose, and timing of exposure relative to the immune stimulus. This review frames this complexity using the recently identified key characteristics of agents that affect immune system function (altered cell signaling, proliferation, differentiation, effector function, communication, trafficking, death, antigen presentation and processing, and tolerance). The use of these key characteristics provides a scaffold for continued discovery of how AHR and its myriad ligands influence the immune system, which will help harness the power of this enigmatic receptor to prevent or treat disease.
Collapse
Affiliation(s)
- Courtney E W Sulentic
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio, USA
| | - Barbara L F Kaplan
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi, USA
| | - B Paige Lawrence
- Department of Environmental Medicine and Department of Microbiology and Immunology, University of Rochester, Rochester, New York, USA;
| |
Collapse
|
3
|
Procházková J, Kahounová Z, Vondráček J, Souček K. Aryl hydrocarbon receptor as a drug target in advanced prostate cancer therapy - obstacles and perspectives. Transcription 2025; 16:47-66. [PMID: 38547312 PMCID: PMC11970783 DOI: 10.1080/21541264.2024.2334106] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 03/08/2024] [Accepted: 03/19/2024] [Indexed: 04/04/2025] Open
Abstract
Aryl hydrocarbon receptor (AhR) is a transcription factor that is primarily known as an intracellular sensor of environmental pollution. After five decades, the list of synthetic and toxic chemicals that activate AhR signaling has been extended to include a number of endogenous compounds produced by various types of cells via their metabolic activity. AhR signaling is active from the very beginning of embryonal development throughout the life cycle and participates in numerous biological processes such as control of cell proliferation and differentiation, metabolism of aromatic compounds of endogenous and exogenous origin, tissue regeneration and stratification, immune system development and polarization, control of stemness potential, and homeostasis maintenance. AhR signaling can be affected by various pharmaceuticals that may help modulate abnormal AhR signaling and drive pathological states. Given their role in immune system development and regulation, AhR antagonistic ligands are attractive candidates for immunotherapy of disease states such as advanced prostate cancer, where an aberrant immune microenvironment contributes to cancer progression and needs to be reeducated. Advanced stages of prostate cancer are therapeutically challenging and characterized by decreased overall survival (OS) due to the metastatic burden. Therefore, this review addresses the role of AhR signaling in the development and progression of prostate cancer and discusses the potential of AhR as a drug target for the treatment of advanced prostate cancer upon entering the phase of drug resistance and failure of first-line androgen deprivation therapy.Abbreviation: ADC: antibody-drug conjugate; ADT: androgen deprivation therapy; AhR: aryl hydrocarbon receptor; AR: androgen receptor; ARE: androgen response element; ARPI: androgen receptor pathway inhibitor; mCRPC: metastatic castration-resistant prostate cancer; DHT: 5a-dihydrotestosterone; FICZ: 6-formylindolo[3,2-b]carbazole; 3-MC: 3-methylcholanthrene; 6-MCDF: 6-methyl-1,3,8-trichlorodibenzofuran; MDSCs: myeloid-derived suppressor cells; PAHs: polycyclic aromatic hydrocarbons; PCa: prostate cancer; TAMs: tumor-associated macrophages; TF: transcription factor; TCDD, 2,3,7,8-tetrachlorodibenzo-p-dioxin; TME: tumor microenvironment; TRAMP: transgenic adenocarcinoma of the mouse prostate; TROP2: tumor associated calcium signal transducer 2.
Collapse
Affiliation(s)
- Jiřina Procházková
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Zuzana Kahounová
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Jan Vondráček
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Karel Souček
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| |
Collapse
|
4
|
Jiang X, Wang J, Lin L, Du L, Ding Y, Zheng F, Xie H, Wang Y, Hu M, Liu B, Xu M, Zhai J, Wang X, Ye J, Cao W, Feng C, Feng J, Hou Z, Meng M, Qiu J, Li Q, Shi Y, Wang Y. Macrophages promote pre-metastatic niche formation of breast cancer through aryl hydrocarbon receptor activity. Signal Transduct Target Ther 2024; 9:352. [PMID: 39690159 DOI: 10.1038/s41392-024-02042-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 10/18/2024] [Accepted: 10/31/2024] [Indexed: 12/19/2024] Open
Abstract
Macrophages that acquire an immunosuppressive phenotype play a crucial role in establishing the pre-metastatic niche (PMN), which is essential for facilitating breast cancer metastasis to distant organs. Our study showed that increased activity of the aryl hydrocarbon receptor (AHR) in lung macrophages plays a crucial role in establishing the immunosuppressive PMN in breast cancer. Specifically, AHR activation led to high expression of PD-L1 on macrophages by directly binding to the promoter of Pdl1. This upregulation of PD-L1 promoted the differentiation of regulatory T cells (Tregs) within the PMN, further enhancing immunosuppressive conditions. Mice with Ahr conditional deletion in macrophages had reduced lung metastasis of breast cancer. The elevated AHR levels in PMN macrophages were induced by GM-CSF, which was secreted by breast cancer cells. Mechanistically, the activated STAT5 signaling pathway induced by GM-CSF prevented AHR from being ubiquitinated, thereby sustaining its activity in macrophages. In breast cancer patients, the expression of AHR and PD-L1 was correlated with increased Treg cell infiltration, and higher levels of AHR were associated with a poor prognosis. These findings reveal that the crosstalk of breast cancer cells, lung macrophages, and Treg cells via the GM-CSF-STAT5-AHR-PD-L1 cascade modulates the lung pre-metastatic niche during breast cancer progression.
Collapse
Affiliation(s)
- Xu Jiang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- The Third Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou, China
| | - Jiaqi Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Liangyu Lin
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Liming Du
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yayun Ding
- The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Fanjun Zheng
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hongzhen Xie
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yu Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Mingyuan Hu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Benming Liu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Muhan Xu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jingjie Zhai
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xuefeng Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jiayin Ye
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wei Cao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Chao Feng
- The Third Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou, China
| | - Jingyi Feng
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zongliu Hou
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan, China
| | - Mingyao Meng
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan, China
| | - Ju Qiu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qing Li
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Yufang Shi
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
- The Third Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou, China.
| | - Ying Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
5
|
Liu S. Aryl Hydrocarbon Receptor Alleviates Hepatic Fibrosis by Inducing Hepatic Stellate Cell Ferroptosis. J Cell Mol Med 2024; 28:e70278. [PMID: 39654034 PMCID: PMC11628353 DOI: 10.1111/jcmm.70278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 11/13/2024] [Accepted: 11/27/2024] [Indexed: 12/12/2024] Open
Abstract
Liver injury-induced activation of hepatic stellate cells (HSCs) is a crucial step in the progression of liver fibrosis. The aryl hydrocarbon receptor (AHR), a ligand-activated transcription factor, is highly expressed in the liver. However, the role of AHR in liver fibrosis remains controversial. Our study revealed that the nontoxic ligand YH439 directly activated the AHR and regulated the expression of multidrug-resistant protein 1 (Mrp1) in mouse hepatic stellate cells (mHSCs), thereby diminishing the antioxidant capacity of mHSCs by promoting GSH efflux, and specifically inducing mHSCs ferroptosis without affecting hepatocytes. In a chronic liver fibrosis model, YH439 activated AHR to promote mHSC ferroptosis without causing hepatocyte ferroptosis, thereby alleviating liver fibrosis. Conclusively, this study shows that AHR alleviates liver fibrosis in mice by selectively inducing mHSC ferroptosis without causing hepatocyte ferroptosis and suggests that AHR is a potential target for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Shenghui Liu
- Lin He's Academician Workstation of New Medicine and Clinical TranslationJining Medical UniversityJiningChina
| |
Collapse
|
6
|
Du X, Tao Q, Fan S, Ren J, Dong Y, Li G, He S, Cao X, Zhu Y. Traditional Mongolian medicine Wu-Lan thirteen-flavor decoction protects rat from hypertension-induced renal injury via aryl hydrocarbon receptor-mediated pathway. Drug Dev Ind Pharm 2024; 50:952-967. [PMID: 39565140 DOI: 10.1080/03639045.2024.2432596] [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: 08/03/2024] [Revised: 11/02/2024] [Accepted: 11/14/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND Wu-Lan Thirteen-Flavour decoction (WLTd), a traditional Mongolian medicine, has been used for treating hypertension in clinical practice, but the chemical basis and underlying mechanisms remain unknown. METHODS The main components of WLTd were identified and quantified using HPLC and UPLC-MS/MS techniques. A compound-target-disease network was constructed using network pharmacology analysis to forecast the potential anti-hypertension targets. In vivo animal and in vitro cellular experiments were performed to validate the efficacy and molecular mechanisms of renal protection of WLTd and its main active components in spontaneous hypertension. RESULTS A total of 136 active compounds in WLTd were collected through relevant databases, and network pharmacology analysis identified that the aryl hydrocarbon receptor (AhR) signaling pathway may serve as a potential anti-hypertension targets. Eight of the active components, including vitexin, kaempferol, toosendanin, ursolic acid, matrine, oxymatrine, gardenoside and quercetin, were identified and quantified by HPLC and UPLC-MS/MS. WLTd effectively lowered the mean blood pressure (159.16 ± 13.91 vs 135 ± 13.37 mmHg), reduced the BUN (391.55 ± 59.96 vs 240.88 ± 51.15 mmol/L) and creatinine (1.78 ± 0.41 vs 0.67 ± 0.34 nmol/L) levels, and reduced hypertension-induced renal damage in SHR. AhR and related key gene expression changes predicted by network pharmacology analysis were validated by immunohistochemistry, RT-qPCR, and Western blot analyses. In vitro, studies also showed that WLTd up-regulated AhR expression in angiotensin II-induced HEK293 cell injury. CONCLUSIONS Wu-Lan Thirteen-Flavour decoction effectively protects hypertension-induced renal injury by regulating the Aryl Hydrocarbon Receptor signaling pathway.
Collapse
Affiliation(s)
- Xiaoli Du
- State Key Laboratory of Component-Based Chinese Medicine and Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Inner Mongolia Key laboratory of Chinese & Mongolian Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Qianqian Tao
- State Key Laboratory of Component-Based Chinese Medicine and Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Siwen Fan
- State Key Laboratory of Component-Based Chinese Medicine and Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jun Ren
- Wuhai Inspection and Testing Center, Wuhai, China
| | - Yu Dong
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| | - Gang Li
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| | - Shuang He
- State Key Laboratory of Component-Based Chinese Medicine and Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaodong Cao
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| | - Yan Zhu
- State Key Laboratory of Component-Based Chinese Medicine and Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
7
|
Mohammadi-Bardbori A, Shadboorestan A, Niknahad H, Noorafshan A, Fardid R, Nadimi E, Bakhtari A, Omidi M. Disrupting Development: Unraveling the Interplay of Aryl Hydrocarbon Receptor (AHR) and Wnt/β-Catenin Pathways in Kidney Development Under the Influence of Environmental Pollutants. Biol Trace Elem Res 2024; 202:4482-4493. [PMID: 38117383 DOI: 10.1007/s12011-023-04009-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 12/08/2023] [Indexed: 12/21/2023]
Abstract
Understanding the intricate molecular mechanisms governing aryl hydrocarbon receptor (AHR) and Wnt/β-Catenin pathways crosstalk is of paramount importance for elucidating normal development. We investigated the repercussions of aberrant activation of these signaling pathways on kidney development. HEK-293 cells were subjected to AHR and Wnt activators and inhibitors for 3 and 24 h. Subsequently, pregnant adult female BALB/c mice were administered treatments at gestation day 9 (GD-9), and embryos were analyzed at GD-18 using a combination of cellular, molecular, stereological, and histopathological techniques. Our results demonstrated a noteworthy escalation in oxidative stress and gene expression endpoints associated with apoptosis. Moreover, stereological analyses exhibited alterations in cortex, proximal tubule, and kidney tissue vessels volumes. Remarkably, co-treatment with 6-formylindolo [3,2-b] carbazole (FICZ) and cadmium (Cd) resulted in a significant reduction in glomerulus volume, while elevating the volumes of distal tubule, Henle loop, and connective tissue, compared to the control group. Histopathological investigations further confirmed structural changes in the loop of Henle and proximal tubule, alongside a decline in glomerular volume. Additionally, the expression levels of AHR and Ctnnb1 genes significantly increased in the Cd-treated group compared to the control group. Enhanced expression of apoptosis-related genes, including Bcl-x, Bax, and Caspase3, along with alterations in mitochondrial membrane potential and cytochrome C release, was observed. In contrast, Gsk3 gene expression was significantly decreased. Our findings robustly establish that chemical pollutants, such as Cd, disrupt the AHR and Wnt/β-Catenin physiological roles during developmental stages by inhibiting the metabolic degradation of FICZ.
Collapse
Affiliation(s)
- Afshin Mohammadi-Bardbori
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Shadboorestan
- Depertment of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Niknahad
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Noorafshan
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Departments of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Fardid
- Department of Radiology, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elham Nadimi
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Azizollah Bakhtari
- Department of Reproductive Biology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahmoud Omidi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| |
Collapse
|
8
|
Pecori Giraldi F, Ferraù F, Ragonese M, Cannavò S. Endocrine disruptors, aryl hydrocarbon receptor and cortisol secretion. J Endocrinol Invest 2024; 47:2407-2419. [PMID: 38637430 PMCID: PMC11393124 DOI: 10.1007/s40618-024-02371-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 03/29/2024] [Indexed: 04/20/2024]
Abstract
PURPOSE Endocrine disruptors exert a plethora of effects in endocrine tissues, from altered function to carcinogenesis. Given its lipophilic nature, the adrenal cortex represents an ideal target for endocrine disruptors and thus, possibly, xenobiotic-induced adrenocortical dysfunction. However, there is no clear understanding of the effect of endocrine disruptors on adrenal steroidogenesis, in particular as regards the aryl hydrocarbon receptor (AHR) pathway, one of the key mediators. METHODS The present review recapitulates available evidence on the effects of AHR ligands on adrenal steroidogenesis, with focus on cortisol secretion. RESULTS Short-term exposure to AHR ligands most often induced a stress-like corticosteroid response followed by decreased responsiveness to stressors with long-term exposure. This was observed in several experimental models across species as well as in animals and humans in real-life settings. Prenatal exposure led to different effects according to sex of the offspring, as observed in murine models and in children from mothers in several countries. In vitro findings proved highly dependent on the experimental setting, with reduced cortisol response and steroidogenic enzyme synthesis mostly observed in fish and increased cortisol synthesis and secretion observed in murine and human adrenal cell lines. Of note, no AHR-binding element was detected in steroidogenic enzyme promoters, suggesting the involvement of additional factors. CONCLUSION Our review provides evidence for the impact of AHR ligands on adrenocortical function and indicates further avenues of research to better clarify its effects.
Collapse
Affiliation(s)
- F Pecori Giraldi
- Department of Clinical Sciences and Community Health, University of Milan, Via Commenda 19, Milan, Italy.
| | - F Ferraù
- Department of Human Pathology of Adulthood and Childhood "Gaetano Barresi,", University of Messina, Messina, Italy
| | - M Ragonese
- Department of Human Pathology of Adulthood and Childhood "Gaetano Barresi,", University of Messina, Messina, Italy
| | - S Cannavò
- Department of Human Pathology of Adulthood and Childhood "Gaetano Barresi,", University of Messina, Messina, Italy
| |
Collapse
|
9
|
Zyoud SH. Global dioxin research trends and focal points: A century-long visual and bibliometric analysis (1923-2022). Toxicol Ind Health 2024; 40:504-518. [PMID: 38838663 DOI: 10.1177/07482337241257276] [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] [Indexed: 06/07/2024]
Abstract
Dioxin-like compounds, recognized by the World Health Organization (WHO) as among the most enduring toxic chemical substances in the environment, are linked to various occupational activities and industrial accidents worldwide. The aim of this study was to examine and present research publications on dioxins, pinpoint current research trends, identify research gaps, and highlight potential avenues for future exploration in the field. The study period for relevant research articles ranged from 1923 to December 31, 2022, and these articles were sourced from the Scopus database. The analysis involved the identification of key contributors to the field and the visualization of topics, themes, and international collaboration. VOSviewer software (version 1.6.20) was used for visualization analysis. A total of 11,620 publications on dioxins were documented in the Scopus database. The predominant category of these documents comprised 9780 original articles, which represents 84.17% of the total publications. The United States lead in the number of publications, with 3992 (34.35%), followed by Japan, with 1429 (12.3%), China, with 1005 (8.65%), and Germany, with 974 (8.38%). Before 2002, scholarly attention in this field focused primarily on the health effects, environmental fate, and mechanism of toxicity of tetrachlorodibenzo-p-dioxin (TCDD). However, a noticeable change in research focus has been observed since 2002, highlighting the emergence of a topic related to the health effects and environmental fate of polychlorinated dibenzo-p-dioxins (PCDDs) and polychlorinated dibenzofurans (PFDFs). This study is the first to conduct a comprehensive quantitative bibliometric analysis of dioxins over time. These findings indicate a significant increase in the overall growth of the dioxin literature over the past 30 years. These findings may prove crucial in guiding and organizing subsequent investigations related to dioxins.
Collapse
Affiliation(s)
- Sa'ed H Zyoud
- Poison Control and Drug Information Center (PCDIC), An-Najah National University, Nablus, Palestine
- Department of Clinical and Community Pharmacy, College of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
- Clinical Research Centre, An-Najah National University Hospital, Nablus, Palestine
| |
Collapse
|
10
|
Zimmerman E, Sturrock A, Reilly CA, Burrell-Gerbers KL, Warren K, Mir-Kasimov M, Zhang MA, Pierce MS, Helms MN, Paine R. Aryl Hydrocarbon Receptor Activation in Pulmonary Alveolar Epithelial Cells Limits Inflammation and Preserves Lung Epithelial Cell Integrity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:600-611. [PMID: 39033086 PMCID: PMC11335325 DOI: 10.4049/jimmunol.2300325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 06/17/2024] [Indexed: 07/23/2024]
Abstract
The aryl hydrocarbon receptor (AHR) is a receptor/transcription factor widely expressed in the lung. The physiological roles of AHR expressed in the alveolar epithelium remain unclear. In this study, we tested the hypothesis that alveolar epithelial AHR activity plays an important role in modulating inflammatory responses and maintaining alveolar integrity during lung injury and repair. AHR is expressed in alveolar epithelial cells (AECs) and is active. AHR activation with the endogenous AHR ligand, FICZ (5,11-dihydroindolo[3,2-b] carbazole-6-carboxaldehyde), significantly suppressed inflammatory cytokine expression in response to inflammatory stimuli in primary murine AECs and in the MLE-15 epithelial cell line. In an LPS model of acute lung injury in mice, coadministration of FICZ with LPS suppressed protein leak, reduced neutrophil accumulation in BAL fluid, and suppressed inflammatory cytokine expression in lung tissue and BAL fluid. Relevant to healing following inflammatory injury, AHR activation suppressed TGF-β-induced expression of genes associated with epithelial-mesenchymal transition. Knockdown of AHR in primary AECs with shRNA or in CRISPR-Cas-9-induced MLE-15 cells resulted in upregulation of α-smooth muscle actin (αSma), Col1a1, and Fn1 and reduced expression of epithelial genes Col4a1 and Sdc1. MLE-15 clones lacking AHR demonstrated accelerated wound closure in a scratch model. AHR activation with FICZ enhanced barrier function (transepithelial electrical resistance) in primary murine AECs and limited decline of transepithelial electrical resistance following inflammatory injury. AHR activation in AECs preserves alveolar integrity by modulating inflammatory cytokine expression while enhancing barrier function and limiting stress-induced expression of mesenchymal genes.
Collapse
Affiliation(s)
- Elizabeth Zimmerman
- Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, University of Utah Spencer Fox Eccles School of Medicine, Salt Lake City, UT
| | - Anne Sturrock
- Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, University of Utah Spencer Fox Eccles School of Medicine, Salt Lake City, UT
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT
| | - Christopher A. Reilly
- Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT
| | | | - Kristi Warren
- Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, University of Utah Spencer Fox Eccles School of Medicine, Salt Lake City, UT
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT
| | - Mustafa Mir-Kasimov
- Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, University of Utah Spencer Fox Eccles School of Medicine, Salt Lake City, UT
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT
| | - Mingyang A. Zhang
- Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, University of Utah Spencer Fox Eccles School of Medicine, Salt Lake City, UT
| | - Megan S. Pierce
- Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, University of Utah Spencer Fox Eccles School of Medicine, Salt Lake City, UT
| | - My N. Helms
- Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, University of Utah Spencer Fox Eccles School of Medicine, Salt Lake City, UT
| | - Robert Paine
- Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, University of Utah Spencer Fox Eccles School of Medicine, Salt Lake City, UT
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT
| |
Collapse
|
11
|
Perrot-Applanat M, Pimpie C, Vacher S, Pocard M, Baud V. High Expression of AhR and Environmental Pollution as AhR-Linked Ligands Impact on Oncogenic Signaling Pathways in Western Patients with Gastric Cancer-A Pilot Study. Biomedicines 2024; 12:1905. [PMID: 39200369 PMCID: PMC11351739 DOI: 10.3390/biomedicines12081905] [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: 06/16/2024] [Revised: 07/29/2024] [Accepted: 08/02/2024] [Indexed: 09/02/2024] Open
Abstract
The vast majority of gastric cancer (GC) cases are adenocarcinomas including intestinal and diffuse GC. The incidence of diffuse GC, often associated with poor overall survival, has constantly increased in Western countries. Epidemiological studies have reported increased mortality from GC after occupational exposure to pro-carcinogens that are metabolically activated by cytochrome P450 enzymes through aryl hydrocarbon receptor (AhR). However, little is known about the role of AhR and environmental AhR ligands in diffuse GC as compared to intestinal GC in Western patients. In a cohort of 29, we demonstrated a significant increase in AhR protein and mRNA expression levels in GCs independently of their subtypes and clinical parameters. AhR and RHOA mRNA expression were correlated in diffuse GC. Further, our study aimed to characterize in GC how AhR and the AhR-related genes cytochrome P450 1A1 (CYP1A1) and P450 1B1 (CYP1B1) affect the mRNA expression of a panel of genes involved in cancer development and progression. In diffuse GC, CYP1A1 expression correlated with genes involved in IGF signaling, epithelial-mesenchymal transition (Vimentin), and migration (MMP2). Using the poorly differentiated KATO III epithelial cell line, two well-known AhR pollutant ligands, namely 2-3-7-8 tetrachlorodibenzo-p-dioxin (TCDD) and benzo[a]pyrene (BaP), strongly increased the expression of CYP1A1 and Interleukin1β (IL1B), and to a lesser extend UGT1, NQO1, and AhR Repressor (AhRR). Moreover, the increased expression of CYP1B1 was seen in diffuse GC, and IHC staining indicated that CYP1B1 is mainly expressed in stromal cells. TCDD treatment increased CYP1B1 expression in KATO III cells, although at lower levels as compared to CYP1A1. In intestinal GC, CYP1B1 expression is inversely correlated with several cancer-related genes such as IDO1, a gene involved in the early steps of tryptophan metabolism that contributes to the endogenous AhR ligand kynurenine expression. Altogether, our data provide evidence for a major role of AhR in GC, as an environmental xenobiotic receptor, through different mechanisms and pathways in diffuse and intestinal GC. Our results support the continued efforts to clarify the identity of exogenous AhR ligands in diffuse GC in order to define new therapeutic strategies.
Collapse
Affiliation(s)
- Martine Perrot-Applanat
- INSERM U1275, Peritoneal Carcimomatosis Paris-Technologies, Hôpital Lariboisiere, Université Paris Cité, 75010 Paris, France; (C.P.); (M.P.)
| | - Cynthia Pimpie
- INSERM U1275, Peritoneal Carcimomatosis Paris-Technologies, Hôpital Lariboisiere, Université Paris Cité, 75010 Paris, France; (C.P.); (M.P.)
| | - Sophie Vacher
- Department of Genetics, Curie Institute, PSL Research University, 75005 Paris, France;
| | - Marc Pocard
- INSERM U1275, Peritoneal Carcimomatosis Paris-Technologies, Hôpital Lariboisiere, Université Paris Cité, 75010 Paris, France; (C.P.); (M.P.)
- Department of Digestive and Oncology Surgery, Hôpital Lariboisiere, Université Paris Cité, 75010 Paris, France
| | - Véronique Baud
- NF-kappaB, Differentiation and Cancer, Faculty of Pharmacy, Université Paris Cité, 75006 Paris, France
| |
Collapse
|
12
|
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
|
13
|
Niu Y, Hu X, Song Y, Wang C, Luo P, Ni S, Jiao F, Qiu J, Jiang W, Yang S, Chen J, Huang R, Jiang H, Chen S, Zhai Q, Xiao J, Guo F. Blautia Coccoides is a Newly Identified Bacterium Increased by Leucine Deprivation and has a Novel Function in Improving Metabolic Disorders. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309255. [PMID: 38429906 PMCID: PMC11095201 DOI: 10.1002/advs.202309255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/14/2024] [Indexed: 03/03/2024]
Abstract
Gut microbiota is linked to human metabolic diseases. The previous work showed that leucine deprivation improved metabolic dysfunction, but whether leucine deprivation alters certain specific species of bacterium that brings these benefits remains unclear. Here, this work finds that leucine deprivation alters gut microbiota composition, which is sufficient and necessary for the metabolic improvements induced by leucine deprivation. Among all the affected bacteria, B. coccoides is markedly increased in the feces of leucine-deprived mice. Moreover, gavage with B. coccoides improves insulin sensitivity and reduces body fat in high-fat diet (HFD) mice, and singly colonization of B. coccoides increases insulin sensitivity in gnotobiotic mice. The effects of B. coccoides are mediated by metabolizing tryptophan into indole-3-acetic acid (I3AA) that activates the aryl hydrocarbon receptor (AhR) in the liver. Finally, this work reveals that reduced fecal B. coccoides and I3AA levels are associated with the clinical metabolic syndrome. These findings suggest that B. coccoides is a newly identified bacterium increased by leucine deprivation, which improves metabolic disorders via metabolizing tryptophan into I3AA.
Collapse
Affiliation(s)
- Yuguo Niu
- Zhongshan HospitalState Key Laboratory of Medical NeurobiologyInstitute for Translational Brain ResearchMOE Frontiers Center for Brain ScienceFudan UniversityShanghai200032China
| | - Xiaoming Hu
- Zhongshan HospitalState Key Laboratory of Medical NeurobiologyInstitute for Translational Brain ResearchMOE Frontiers Center for Brain ScienceFudan UniversityShanghai200032China
| | - Yali Song
- Department of Metabolic and Bariatric Surgery and Clinical Research InstituteFirst Affiliated Hospital of Jinan UniversityGuangzhou510632China
| | - Cunchuan Wang
- Department of Metabolic and Bariatric Surgery and Clinical Research InstituteFirst Affiliated Hospital of Jinan UniversityGuangzhou510632China
| | - Peixiang Luo
- CAS Key Laboratory of NutritionMetabolism and Food SafetyInnovation Center for Intervention of Chronic Disease and Promotion of HealthShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
| | - Shihong Ni
- Zhongshan HospitalState Key Laboratory of Medical NeurobiologyInstitute for Translational Brain ResearchMOE Frontiers Center for Brain ScienceFudan UniversityShanghai200032China
| | - Fuxin Jiao
- CAS Key Laboratory of NutritionMetabolism and Food SafetyInnovation Center for Intervention of Chronic Disease and Promotion of HealthShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
| | - Ju Qiu
- CAS Key Laboratory of NutritionMetabolism and Food SafetyInnovation Center for Intervention of Chronic Disease and Promotion of HealthShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
| | - Weihong Jiang
- Key Laboratory of Synthetic BiologyInstitute of Plant Physiology and EcologyCAS Center for Excellence in Molecular Plant ScienceShanghai200032China
| | - Sheng Yang
- Key Laboratory of Synthetic BiologyInstitute of Plant Physiology and EcologyCAS Center for Excellence in Molecular Plant ScienceShanghai200032China
| | - Jun Chen
- Key Laboratory of Synthetic BiologyInstitute of Plant Physiology and EcologyCAS Center for Excellence in Molecular Plant ScienceShanghai200032China
| | - Rui Huang
- CAS Key Laboratory of NutritionMetabolism and Food SafetyInnovation Center for Intervention of Chronic Disease and Promotion of HealthShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
| | - Haizhou Jiang
- Zhongshan HospitalState Key Laboratory of Medical NeurobiologyInstitute for Translational Brain ResearchMOE Frontiers Center for Brain ScienceFudan UniversityShanghai200032China
| | - Shanghai Chen
- Zhongshan HospitalState Key Laboratory of Medical NeurobiologyInstitute for Translational Brain ResearchMOE Frontiers Center for Brain ScienceFudan UniversityShanghai200032China
| | - Qiwei Zhai
- CAS Key Laboratory of NutritionMetabolism and Food SafetyInnovation Center for Intervention of Chronic Disease and Promotion of HealthShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
| | - Jia Xiao
- Department of Metabolic and Bariatric Surgery and Clinical Research InstituteFirst Affiliated Hospital of Jinan UniversityGuangzhou510632China
| | - Feifan Guo
- Zhongshan HospitalState Key Laboratory of Medical NeurobiologyInstitute for Translational Brain ResearchMOE Frontiers Center for Brain ScienceFudan UniversityShanghai200032China
| |
Collapse
|
14
|
Rodrigues SG, van der Merwe S, Krag A, Wiest R. Gut-liver axis: Pathophysiological concepts and medical perspective in chronic liver diseases. Semin Immunol 2024; 71:101859. [PMID: 38219459 DOI: 10.1016/j.smim.2023.101859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 10/11/2023] [Accepted: 12/04/2023] [Indexed: 01/16/2024]
Affiliation(s)
- Susana G Rodrigues
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Schalk van der Merwe
- Department of Gastroenterology and Hepatology, University hospital Gasthuisberg, University of Leuven, Belgium
| | - Aleksander Krag
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark; Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark, University of Southern Denmark, Odense, Denmark
| | - Reiner Wiest
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland.
| |
Collapse
|
15
|
Nebert DW. Gene-Environment Interactions: My Unique Journey. Annu Rev Pharmacol Toxicol 2024; 64:1-26. [PMID: 37788491 DOI: 10.1146/annurev-pharmtox-022323-082311] [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] [Indexed: 10/05/2023]
Abstract
I am deeply honored to be invited to write this scientific autobiography. As a physician-scientist, pediatrician, molecular biologist, and geneticist, I have authored/coauthored more than 600 publications in the fields of clinical medicine, biochemistry, biophysics, pharmacology, drug metabolism, toxicology, molecular biology, cancer, standardized gene nomenclature, developmental toxicology and teratogenesis, mouse genetics, human genetics, and evolutionary genomics. Looking back, I think my career can be divided into four distinct research areas, which I summarize mostly chronologically in this article: (a) discovery and characterization of the AHR/CYP1 axis, (b) pharmacogenomics and genetic prediction of response to drugs and other environmental toxicants, (c) standardized drug-metabolizing gene nomenclature based on evolutionary divergence, and (d) discovery and characterization of the SLC39A8 gene encoding the ZIP8 metal cation influx transporter. Collectively, all four topics embrace gene-environment interactions, hence the title of my autobiography.
Collapse
Affiliation(s)
- Daniel W Nebert
- Department of Environmental and Public Health Sciences and Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Department of Pediatrics and Molecular Developmental Biology, Division of Human Genetics, Cincinnati Children's Hospital, Cincinnati, Ohio, USA;
| |
Collapse
|
16
|
Trujillo-Ochoa JL, Kazemian M, Afzali B. The role of transcription factors in shaping regulatory T cell identity. Nat Rev Immunol 2023; 23:842-856. [PMID: 37336954 PMCID: PMC10893967 DOI: 10.1038/s41577-023-00893-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2023] [Indexed: 06/21/2023]
Abstract
Forkhead box protein 3-expressing (FOXP3+) regulatory T cells (Treg cells) suppress conventional T cells and are essential for immunological tolerance. FOXP3, the master transcription factor of Treg cells, controls the expression of multiples genes to guide Treg cell differentiation and function. However, only a small fraction (<10%) of Treg cell-associated genes are directly bound by FOXP3, and FOXP3 alone is insufficient to fully specify the Treg cell programme, indicating a role for other accessory transcription factors operating upstream, downstream and/or concurrently with FOXP3 to direct Treg cell specification and specialized functions. Indeed, the heterogeneity of Treg cells can be at least partially attributed to differential expression of transcription factors that fine-tune their trafficking, survival and functional properties, some of which are niche-specific. In this Review, we discuss the emerging roles of accessory transcription factors in controlling Treg cell identity. We specifically focus on members of the basic helix-loop-helix family (AHR), basic leucine zipper family (BACH2, NFIL3 and BATF), CUT homeobox family (SATB1), zinc-finger domain family (BLIMP1, Ikaros and BCL-11B) and interferon regulatory factor family (IRF4), as well as lineage-defining transcription factors (T-bet, GATA3, RORγt and BCL-6). Understanding the imprinting of Treg cell identity and specialized function will be key to unravelling basic mechanisms of autoimmunity and identifying novel targets for drug development.
Collapse
Affiliation(s)
- Jorge L Trujillo-Ochoa
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA
| | - Majid Kazemian
- Departments of Biochemistry and Computer Science, Purdue University, West Lafayette, IN, USA
| | - Behdad Afzali
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA.
| |
Collapse
|
17
|
He Y, Liu W, Gao L, Ren Z, Hussain J, Jia T, Mao T, Deng J, Xu X, Yin F. Occurrence and Formation Mechanism of PCDD/Fs and SCCPs in Chlorinated Paraffin Products. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:17522-17533. [PMID: 37905521 DOI: 10.1021/acs.est.3c06378] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Polychlorinated dibenzo-p-dioxins and dibenzofurans (PCDD/Fs) and short-chain chlorinated paraffins (SCCPs) can be formed during the production of chlorinated paraffins (CPs). Detection and accurate quantification of PCDD/Fs in CPs are challenging because of their matrix complexity. Therefore, the occurrence and formation mechanisms of PCDD/Fs from CPs have not been studied extensively in the past. In this study, 15 commercial samples including solid and liquid CPs were collected in 2022 from China. The average ΣSCCP concentrations detected in the solid and liquid CPs were 158 and 137 mg/g, respectively. The average International Toxic Equivalent (I-TEQ) values of 2,3,7,8-PCDD/F in solid and liquid CPs were 15.8 pg I-TEQ/g and 15.0 pg I-TEQ/g, respectively. The solid and liquid CPs had different predominant congener groups for SCCPs and PCDD/Fs. Possible formation routes for the generation of PCDD/Fs were analyzed by screening precursors in paraffin and laboratory-scale thermochemical experiments of CPs. The transformation between 2,3,7,8-PCDD/Fs and non-2,3,7,8-PCDD/Fs was recognized by calculating the successive chlorination preference. The first reported occurrence of PCDD/Fs in CP commercial products indicated that exposure to CPs and downstream products might be an assignable source of PCDD/F emission, which is of great significance to further explore the control factors of PCDD/Fs in the whole life cycle of CPs.
Collapse
Affiliation(s)
- Yunchen He
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 101314, China
| | - Wenbin Liu
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 101314, China
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Lirong Gao
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 101314, China
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Zhiyuan Ren
- Foreign Environmental Cooperation Center, Ministry of Ecology and Environment, Beijing 100035, China
| | - Javid Hussain
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 101314, China
- Department of Environmental Sciences, Balochistan University of Information Technology, Engineering and Management Sciences, Quetta 87100, Pakistan
| | - Tianqi Jia
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 101314, China
| | - Tianao Mao
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 101314, China
| | - Jinglin Deng
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 101314, China
| | - Xiaotian Xu
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Fei Yin
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 101314, China
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| |
Collapse
|
18
|
Tumova S, Dolezel D, Jindra M. Conserved and Unique Roles of bHLH-PAS Transcription Factors in Insects - From Clock to Hormone Reception. J Mol Biol 2023; 436:168332. [PMID: 39491146 DOI: 10.1016/j.jmb.2023.168332] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/05/2024]
Abstract
A dozen bHLH-PAS transcription factors have evolved since the dawn of the animal kingdom; nine of them have mutual orthologs between arthropods and vertebrates. These proteins are master regulators in a range of developmental processes from organogenesis, nervous system formation and functioning, to cell fate decisions defining identity of limbs or photoreceptors for color vision. Among the functionally best conserved are bHLH-PAS proteins acting in the animal circadian clock. On the other side of the spectrum are fundamental physiological mechanisms such as those underlying xenobiotic detoxification, oxygen homeostasis, and metabolic adaptation to hypoxia, infection or tumor progression. Predictably, malfunctioning of bHLH-PAS regulators leads to pathologies. Performance of the individual bHLH-PAS proteins is modulated at multiple levels including dimerization and other protein-protein interactions, proteasomal degradation, and by binding low-molecular weight ligands. Despite the vast evolutionary gap dividing arthropods and vertebrates, and the differences in their anatomy, many functions of orthologous bHLH-PAS proteins are remarkably similar, including at the molecular level. Our phylogenetic analysis shows that one bHLH-PAS protein type has been lost during vertebrate evolution. This protein has a unique function as a receptor of the sesquiterpenoid juvenile hormones of insects and crustaceans. Although some other bHLH-PAS proteins are regulated by binding small molecules, the juvenile hormone receptor presents an unprecedented case, since all other non-peptide animal hormones activate members of the nuclear receptor family. The purpose of this review is to compare and highlight parallels and differences in functioning of bHLH-PAS proteins between insects and vertebrates.
Collapse
Affiliation(s)
- Sarka Tumova
- Institute of Entomology, Biology Center of the Czech Academy of Sciences, Ceske Budejovice 37005, Czech Republic
| | - David Dolezel
- Institute of Entomology, Biology Center of the Czech Academy of Sciences, Ceske Budejovice 37005, Czech Republic
| | - Marek Jindra
- Institute of Entomology, Biology Center of the Czech Academy of Sciences, Ceske Budejovice 37005, Czech Republic.
| |
Collapse
|
19
|
Hou JJ, Ma AH, Qin YH. Activation of the aryl hydrocarbon receptor in inflammatory bowel disease: insights from gut microbiota. Front Cell Infect Microbiol 2023; 13:1279172. [PMID: 37942478 PMCID: PMC10628454 DOI: 10.3389/fcimb.2023.1279172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/09/2023] [Indexed: 11/10/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory intestinal disease that affects more than 3.5 million people, with rising prevalence. It deeply affects patients' daily life, increasing the burden on patients, families, and society. Presently, the etiology of IBD remains incompletely clarified, while emerging evidence has demonstrated that altered gut microbiota and decreased aryl hydrocarbon receptor (AHR) activity are closely associated with IBD. Furthermore, microbial metabolites are capable of AHR activation as AHR ligands, while the AHR, in turn, affects the microbiota through various pathways. In light of the complex connection among gut microbiota, the AHR, and IBD, it is urgent to review the latest research progress in this field. In this review, we describe the role of gut microbiota and AHR activation in IBD and discussed the crosstalk between gut microbiota and the AHR in the context of IBD. Taken as a whole, we propose new therapeutic strategies targeting the AHR-microbiota axis for IBD, even for other related diseases caused by AHR-microbiota dysbiosis.
Collapse
Affiliation(s)
| | | | - Yue-Hua Qin
- Department of Gastroenterology, Shaoxing People’s Hospital, Shaoxing, China
| |
Collapse
|
20
|
Xiong L, Helm EY, Dean JW, Sun N, Jimenez-Rondan FR, Zhou L. Nutrition impact on ILC3 maintenance and function centers on a cell-intrinsic CD71-iron axis. Nat Immunol 2023; 24:1671-1684. [PMID: 37709985 PMCID: PMC11256193 DOI: 10.1038/s41590-023-01612-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 08/04/2023] [Indexed: 09/16/2023]
Abstract
Iron metabolism is pivotal for cell fitness in the mammalian host; however, its role in group 3 innate lymphoid cells (ILC3s) is unknown. Here we show that transferrin receptor CD71 (encoded by Tfrc)-mediated iron metabolism cell-intrinsically controls ILC3 proliferation and host protection against Citrobacter rodentium infection and metabolically affects mitochondrial respiration by switching of oxidative phosphorylation toward glycolysis. Iron deprivation or Tfrc ablation in ILC3s reduces the expression and/or activity of the aryl hydrocarbon receptor (Ahr), a key ILC3 regulator. Genetic ablation or activation of Ahr in ILC3s leads to CD71 upregulation or downregulation, respectively, suggesting Ahr-mediated suppression of CD71. Mechanistically, Ahr directly binds to the Tfrc promoter to inhibit transcription. Iron overload partially restores the defective ILC3 compartment in the small intestine of Ahr-deficient mice, consistent with the compensatory upregulation of CD71. These data collectively demonstrate an under-appreciated role of the Ahr-CD71-iron axis in the regulation of ILC3 maintenance and function.
Collapse
Affiliation(s)
- Lifeng Xiong
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Eric Y Helm
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Joseph W Dean
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Na Sun
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Felix R Jimenez-Rondan
- Center for Nutritional Sciences and Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, USA
| | - Liang Zhou
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
21
|
Huang Z, de Vries S, Fogliano V, Wells JM, van der Wielen N, Capuano E. Effect of whole foods on the microbial production of tryptophan-derived aryl hydrocarbon receptor agonists in growing pigs. Food Chem 2023; 416:135804. [PMID: 36893645 DOI: 10.1016/j.foodchem.2023.135804] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/27/2023] [Accepted: 02/24/2023] [Indexed: 03/05/2023]
Abstract
Effects of whole foods on the microbial production of tryptophan-derived aryl hydrocarbon receptor (AhR) ligands in the intestine were investigated in a pig model. Ileal digesta and faeces of pigs after feeding of eighteen different foods were analyzed. Indole, indole-3-propionic acid, indole-3-acetic acid, indole-3-lactic acid, kynurenine, tryptamine, and indole-3-aldehyde were identified in ileal digesta, which were also identified in faeces but at higher concentrations except indole-3-lactic acid, together with skatole, oxindole, serotonin, and indoleacrylic acid. The panel of tryptophan catabolites in ileal digesta and faeces varied across different foods. Eggs induced the highest overall concentration of catabolites in ileal digesta dominated by indole. Amaranth induced the highest overall concentration of catabolites in faeces dominated by skatole. Using a reporter cell line, we observed many faecal samples but not ileal samples retained AhR activity. Collectively, these findings contribute to food selection targeting AhR ligands production from dietary tryptophan in the intestine.
Collapse
Affiliation(s)
- Zhan Huang
- Food Quality and Design Group, Department of Agrotechnology and Food Sciences, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, The Netherlands; Host-Microbe Interactomics Group, Department of Animal Sciences, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, The Netherlands.
| | - Sonja de Vries
- Animal Nutrition Group, Department of Animal Sciences, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, The Netherlands
| | - Vincenzo Fogliano
- Food Quality and Design Group, Department of Agrotechnology and Food Sciences, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, The Netherlands
| | - Jerry M Wells
- Host-Microbe Interactomics Group, Department of Animal Sciences, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, The Netherlands
| | - Nikkie van der Wielen
- Animal Nutrition Group, Department of Animal Sciences, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, The Netherlands; Division of Human Nutrition and Health, Department of Agrotechnology and Food Sciences, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, The Netherlands
| | - Edoardo Capuano
- Food Quality and Design Group, Department of Agrotechnology and Food Sciences, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, The Netherlands.
| |
Collapse
|
22
|
Riddick DS. Fifty Years of Aryl Hydrocarbon Receptor Research as Reflected in the Pages of Drug Metabolism and Disposition. Drug Metab Dispos 2023; 51:657-671. [PMID: 36653119 DOI: 10.1124/dmd.122.001009] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 01/19/2023] Open
Abstract
The induction of multiple drug-metabolizing enzymes by halogenated and polycyclic aromatic hydrocarbon toxicants is mediated by the aryl hydrocarbon receptor (AHR). This fascinating receptor also has natural dietary and endogenous ligands, and much is now appreciated about the AHR's developmental and physiologic roles, as well as its importance in cancer and other diseases. The past several years has witnessed increasing emphasis on understanding the multifaceted roles of the AHR in the immune system. Most would agree that the "discovery" of the AHR occurred in 1976, with the report of specific binding of a high affinity radioligand in mouse liver, just three years after the launch of the journal Drug Metabolism and Disposition (DMD) in 1973. Over the ensuing 50 years, the AHR and DMD have led parallel and often intersecting lives. The overall goal of this mini-review is to provide a decade-by-decade overview of major historical landmark discoveries in the AHR field and to highlight the numerous contributions made by publications appearing in the pages of DMD. It is hoped that this historical tour might inspire current and future research in the AHR field. SIGNIFICANCE STATEMENT: With the launch of Drug Metabolism and Disposition (DMD) in 1973 and the discovery of the aryl hydrocarbon receptor (AHR) in 1976, the journal and the receptor have led parallel and often intersecting lives over the past 50 years. Tracing the history of the AHR can reveal how knowledge in the field has evolved to the present and highlight the important contributions made by discoveries reported in DMD. This may inspire additional DMD papers reporting future AHR landmark discoveries.
Collapse
Affiliation(s)
- David S Riddick
- Department of Pharmacology and Toxicology, Medical Sciences Building, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
23
|
Heo MJ, Suh JH, Lee SH, Poulsen KL, An YA, Moorthy B, Hartig SM, Moore DD, Kim KH. Aryl hydrocarbon receptor maintains hepatic mitochondrial homeostasis in mice. Mol Metab 2023; 72:101717. [PMID: 37004989 PMCID: PMC10106517 DOI: 10.1016/j.molmet.2023.101717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/09/2023] [Accepted: 03/26/2023] [Indexed: 04/03/2023] Open
Abstract
OBJECTIVE Mitophagy removes damaged mitochondria to maintain cellular homeostasis. Aryl hydrocarbon receptor (AhR) expression in the liver plays a crucial role in supporting normal liver functions, but its impact on mitochondrial function is unclear. Here, we identified a new role of AhR in the regulation of mitophagy to control hepatic energy homeostasis. METHODS In this study, we utilized primary hepatocytes from AhR knockout (KO) mice and AhR knockdown AML12 hepatocytes. An endogenous AhR ligand, kynurenine (Kyn), was used to activate AhR in AML12 hepatocytes. Mitochondrial function and mitophagy process were comprehensively assessed by MitoSOX and mt-Keima fluorescence imaging, Seahorse XF-based oxygen consumption rate measurement, and Mitoplate S-1 mitochondrial substrate utilization analysis. RESULTS Transcriptomic analysis indicated that mitochondria-related gene sets were dysregulated in AhR KO liver. In both primary mouse hepatocytes and AML12 hepatocyte cell lines, AhR inhibition strongly suppressed mitochondrial respiration rate and substrate utilization. AhR inhibition also blunted the fasting response of several essential autophagy genes and the mitophagy process. We further identified BCL2 interacting protein 3 (BNIP3), a mitophagy receptor that senses nutrient stress, as an AhR target gene. AhR is directly recruited to the Bnip3 genomic locus, and Bnip3 transcription was enhanced by AhR endogenous ligand treatment in wild-type liver and abolished entirely in AhR KO liver. Mechanistically, overexpression of Bnip3 in AhR knockdown cells mitigated the production of mitochondrial reactive oxygen species (ROS) and restored functional mitophagy. CONCLUSIONS AhR regulation of the mitophagy receptor BNIP3 coordinates hepatic mitochondrial function. Loss of AhR induces mitochondrial ROS production and impairs mitochondrial respiration. These findings provide new insight into how endogenous AhR governs hepatic mitochondrial homeostasis.
Collapse
Affiliation(s)
- Mi Jeong Heo
- Department of Anesthesiology, Critical Care and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Ji Ho Suh
- Department of Anesthesiology, Critical Care and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Sung Ho Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju 61186, South Korea
| | - Kyle L Poulsen
- Department of Anesthesiology, Critical Care and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yu A An
- Department of Anesthesiology, Critical Care and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Bhagavatula Moorthy
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Texas Children's Hospital, Houston, TX 77030, USA
| | - Sean M Hartig
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - David D Moore
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA.
| | - Kang Ho Kim
- Department of Anesthesiology, Critical Care and Pain Medicine and Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
24
|
Chen Y, Wang Y, Fu Y, Yin Y, Xu K. Modulating AHR function offers exciting therapeutic potential in gut immunity and inflammation. Cell Biosci 2023; 13:85. [PMID: 37179416 PMCID: PMC10182712 DOI: 10.1186/s13578-023-01046-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
Aryl hydrocarbon receptor (AHR) is a ligand-dependent transcription factor. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is a classical exogenous synthetic ligand of AHR that has significant immunotoxic effects. Activation of AHR has beneficial effects on intestinal immune responses, but inactivation or overactivation of AHR can lead to intestinal immune dysregulation and even intestinal diseases. Sustained potent activation of AHR by TCDD results in impairment of the intestinal epithelial barrier. However, currently, AHR research has been more focused on elucidating physiologic AHR function than on dioxin toxicity. The appropriate level of AHR activation plays a role in maintaining gut health and protecting against intestinal inflammation. Therefore, AHR offers a crucial target to modulate intestinal immunity and inflammation. Herein, we summarize our current understanding of the relationship between AHR and intestinal immunity, the ways in which AHR affects intestinal immunity and inflammation, the effects of AHR activity on intestinal immunity and inflammation, and the effect of dietary habits on intestinal health through AHR. Finally, we discuss the therapeutic role of AHR in maintaining gut homeostasis and relieving inflammation.
Collapse
Affiliation(s)
- Yue Chen
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450000, China
| | - Yadong Wang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Yawei Fu
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450000, China
| | - Yulong Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450000, China
| | - Kang Xu
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
| |
Collapse
|
25
|
Pinto CJG, Ávila-Gálvez MÁ, Lian Y, Moura-Alves P, Nunes Dos Santos C. Targeting the aryl hydrocarbon receptor by gut phenolic metabolites: A strategy towards gut inflammation. Redox Biol 2023; 61:102622. [PMID: 36812782 PMCID: PMC9958510 DOI: 10.1016/j.redox.2023.102622] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 01/25/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023] Open
Abstract
The Aryl Hydrocarbon Receptor (AHR) is a ligand-dependent transcription factor able to control complex transcriptional processes in several cell types, which has been correlated with various diseases, including inflammatory bowel diseases (IBD). Numerous studies have described different compounds as ligands of this receptor, like xenobiotics, natural compounds, and several host-derived metabolites. Dietary (poly)phenols have been studied regarding their pleiotropic activities (e.g., neuroprotective and anti-inflammatory), but their AHR modulatory capabilities have also been considered. However, dietary (poly)phenols are submitted to extensive metabolism in the gut (e.g., gut microbiota). Thus, the resulting gut phenolic metabolites could be key players modulating AHR since they are the ones that reach the cells and may exert effects on the AHR throughout the gut and other organs. This review aims at a comprehensive search for the most abundant gut phenolic metabolites detected and quantified in humans to understand how many have been described as AHR modulators and what could be their impact on inflammatory gut processes. Even though several phenolic compounds have been studied regarding their anti-inflammatory capacities, only 1 gut phenolic metabolite, described as AHR modulator, has been evaluated on intestinal inflammatory models. Searching for AHR ligands could be a novel strategy against IBD.
Collapse
Affiliation(s)
- Catarina J G Pinto
- iNOVA4Health, NOVA Medical School
- Faculdade de Ciências Médicas, NMS
- FCM, Universidade Nova de Lisboa, Lisboa, Portugal; IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal; I3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | - María Ángeles Ávila-Gálvez
- iNOVA4Health, NOVA Medical School
- Faculdade de Ciências Médicas, NMS
- FCM, Universidade Nova de Lisboa, Lisboa, Portugal; iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras, Portugal
| | - Yilong Lian
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, OX3 7DQ, Oxford, United Kingdom
| | - Pedro Moura-Alves
- IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal; I3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal; Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, OX3 7DQ, Oxford, United Kingdom.
| | - Cláudia Nunes Dos Santos
- iNOVA4Health, NOVA Medical School
- Faculdade de Ciências Médicas, NMS
- FCM, Universidade Nova de Lisboa, Lisboa, Portugal; iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, Oeiras, Portugal.
| |
Collapse
|
26
|
Salminen A. Aryl hydrocarbon receptor (AhR) impairs circadian regulation: impact on the aging process. Ageing Res Rev 2023; 87:101928. [PMID: 37031728 DOI: 10.1016/j.arr.2023.101928] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/23/2023] [Accepted: 04/06/2023] [Indexed: 04/11/2023]
Abstract
Circadian clocks control the internal sleep-wake rhythmicity of 24hours which is synchronized by the solar cycle. Circadian regulation of metabolism evolved about 2.5 billion years ago, i.e., the rhythmicity has been conserved from cyanobacteria and Archaea through to mammals although the mechanisms utilized have developed with evolution. While the aryl hydrocarbon receptor (AhR) is an evolutionarily conserved defence mechanism against environmental threats, it has gained many novel functions during evolution, such as the regulation of cell cycle, proteostasis, and many immune functions. There is robust evidence that AhR signaling impairs circadian rhythmicity, e.g., by interacting with the core BMAL1/CLOCK complex and disturbing the epigenetic regulation of clock genes. The maintenance of circadian rhythms is impaired with aging, disturbing metabolism and many important functions in aged organisms. Interestingly, it is known that AhR signaling promotes an age-related tissue degeneration, e.g., it is able to inhibit autophagy, enhance cellular senescence, and disrupt extracellular matrix. These alterations are rather similar to those induced by a long-term impairment of circadian rhythms. However, it is not known whether AhR signaling enhances the aging process by impairing circadian homeostasis. I will examine the experimental evidence indicating that AhR signaling is able to promote the age-related degeneration via a disruption of circadian rhythmicity.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| |
Collapse
|
27
|
Elson DJ, Kolluri SK. Tumor-Suppressive Functions of the Aryl Hydrocarbon Receptor (AhR) and AhR as a Therapeutic Target in Cancer. BIOLOGY 2023; 12:526. [PMID: 37106727 PMCID: PMC10135996 DOI: 10.3390/biology12040526] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/25/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor involved in regulating a wide range of biological responses. A diverse array of xenobiotics and endogenous small molecules bind to the receptor and drive unique phenotypic responses. Due in part to its role in mediating toxic responses to environmental pollutants, AhR activation has not been traditionally viewed as a viable therapeutic approach. Nonetheless, the expression and activation of AhR can inhibit the proliferation, migration, and survival of cancer cells, and many clinically approved drugs transcriptionally activate AhR. Identification of novel select modulators of AhR-regulated transcription that promote tumor suppression is an active area of investigation. The development of AhR-targeted anticancer agents requires a thorough understanding of the molecular mechanisms driving tumor suppression. Here, we summarized the tumor-suppressive mechanisms regulated by AhR with an emphasis on the endogenous functions of the receptor in opposing carcinogenesis. In multiple different cancer models, the deletion of AhR promotes increased tumorigenesis, but a precise understanding of the molecular cues and the genetic targets of AhR involved in this process is lacking. The intent of this review was to synthesize the evidence supporting AhR-dependent tumor suppression and distill insights for development of AhR-targeted cancer therapeutics.
Collapse
Affiliation(s)
- Daniel J. Elson
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA
| | - Siva K. Kolluri
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
| |
Collapse
|
28
|
Perdew GH, Esser C, Snyder M, Sherr DH, van den Bogaard EH, McGovern K, Fernández-Salguero PM, Coumoul X, Patterson AD. The Ah Receptor from Toxicity to Therapeutics: Report from the 5th AHR Meeting at Penn State University, USA, June 2022. Int J Mol Sci 2023; 24:5550. [PMID: 36982624 PMCID: PMC10058801 DOI: 10.3390/ijms24065550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/10/2023] [Indexed: 03/16/2023] Open
Abstract
The aryl hydrocarbon receptor (AHR) is a sensor of low-molecular-weight molecule signals that originate from environmental exposures, the microbiome, and host metabolism. Building upon initial studies examining anthropogenic chemical exposures, the list of AHR ligands of microbial, diet, and host metabolism origin continues to grow and has provided important clues as to the function of this enigmatic receptor. The AHR has now been shown to be directly involved in numerous biochemical pathways that influence host homeostasis, chronic disease development, and responses to toxic insults. As this field of study has continued to grow, it has become apparent that the AHR is an important novel target for cancer, metabolic diseases, skin conditions, and autoimmune disease. This meeting attempted to cover the scope of basic and applied research being performed to address possible applications of our basic knowledge of this receptor on therapeutic outcomes.
Collapse
Affiliation(s)
- Gary H. Perdew
- Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, Penn State University, University Park, PA 16802, USA
| | - Charlotte Esser
- IUF-Leibniz Research Institute for Environmental Medicine, Auf’m Hennekamp 50, 40225 Düsseldorf, Germany
| | - Megan Snyder
- Department of Environmental Health, Boston University School of Public Health, 72 East Concord Street, Boston, MA 02118, USA
| | - David H. Sherr
- Department of Environmental Health, Boston University School of Public Health, 72 East Concord Street, Boston, MA 02118, USA
| | - Ellen H. van den Bogaard
- Department of Dermatology, Radboud University Medical Center, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Karen McGovern
- Ikena Oncology, Inc., 645 Summer Street Suite 101, Boston, MA 02210, USA
| | - Pedro M. Fernández-Salguero
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, Universidad de Extremadura, Avenida de Elvas s/n, 06071 Badajoz, Spain
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), Avenida de la Investigación s/n, 06071 Badajoz, Spain
| | - Xavier Coumoul
- INSERM UMR-S1124, 45 rue des Saints-Peères, 75006 Paris, France
| | - Andrew D. Patterson
- Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, Penn State University, University Park, PA 16802, USA
| |
Collapse
|
29
|
Patil NY, Friedman JE, Joshi AD. Role of Hepatic Aryl Hydrocarbon Receptor in Non-Alcoholic Fatty Liver Disease. RECEPTORS (BASEL, SWITZERLAND) 2023; 2:1-15. [PMID: 37284280 PMCID: PMC10240927 DOI: 10.3390/receptors2010001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Numerous nuclear receptors including farnesoid X receptor, liver X receptor, peroxisome proliferator-activated receptors, pregnane X receptor, hepatic nuclear factors have been extensively studied within the context of non-alcoholic fatty liver disease (NAFLD). Following the first description of the Aryl hydrocarbon Receptor (AhR) in the 1970s and decades of research which unveiled its role in toxicity and pathophysiological processes, the functional significance of AhR in NAFLD has not been completely decoded. Recently, multiple research groups have utilized a plethora of in vitro and in vivo models that mimic NAFLD pathology to investigate the functional significance of AhR in fatty liver disease. This review provides a comprehensive account of studies describing both the beneficial and possible detrimental role of AhR in NAFLD. A plausible reconciliation for the paradox indicating AhR as a 'double-edged sword' in NAFLD is discussed. Finally, understanding AhR ligands and their signaling in NAFLD will facilitate us to probe AhR as a potential drug target to design innovative therapeutics against NAFLD in the near future.
Collapse
Affiliation(s)
- Nikhil Y. Patil
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
| | - Jacob E. Friedman
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
| | - Aditya D. Joshi
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
| |
Collapse
|
30
|
Silva-Parra J, Sandu C, Felder-Schmittbuhl MP, Hernández-Kelly LC, Ortega A. Aryl Hydrocarbon Receptor in Glia Cells: A Plausible Glutamatergic Neurotransmission Orchestrator. Neurotox Res 2023; 41:103-117. [PMID: 36607593 DOI: 10.1007/s12640-022-00623-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/23/2022] [Accepted: 12/15/2022] [Indexed: 01/07/2023]
Abstract
Glutamate is the major excitatory amino acid in the vertebrate brain. Glutamatergic signaling is involved in most of the central nervous system functions. Its main components, namely receptors, ion channels, and transporters, are tightly regulated at the transcriptional, translational, and post-translational levels through a diverse array of extracellular signals, such as food, light, and neuroactive molecules. An exquisite and well-coordinated glial/neuronal bidirectional communication is required for proper excitatory amino acid signal transactions. Biochemical shuttles such as the glutamate/glutamine and the astrocyte-neuronal lactate represent the fundamental involvement of glial cells in glutamatergic transmission. In fact, the disruption of any of these coordinated biochemical intercellular cascades leads to an excitotoxic insult that underlies some aspects of most of the neurodegenerative diseases characterized thus far. In this contribution, we provide a comprehensive summary of the involvement of the Aryl hydrocarbon receptor, a ligand-dependent transcription factor in the gene expression regulation of glial glutamate transporters. These receptors might serve as potential targets for the development of novel strategies for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Janisse Silva-Parra
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, San Pedro Zacatenco, 07360, CDMX, México
| | - Cristina Sandu
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, Strasbourg, France
| | - Marie-Paule Felder-Schmittbuhl
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, Strasbourg, France
| | - Luisa C Hernández-Kelly
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, San Pedro Zacatenco, 07360, CDMX, México
| | - Arturo Ortega
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, San Pedro Zacatenco, 07360, CDMX, México.
| |
Collapse
|
31
|
Dean JW, Helm EY, Fu Z, Xiong L, Sun N, Oliff KN, Muehlbauer M, Avram D, Zhou L. The aryl hydrocarbon receptor cell intrinsically promotes resident memory CD8 + T cell differentiation and function. Cell Rep 2023; 42:111963. [PMID: 36640340 PMCID: PMC9940759 DOI: 10.1016/j.celrep.2022.111963] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 11/23/2022] [Accepted: 12/19/2022] [Indexed: 01/06/2023] Open
Abstract
The Aryl hydrocarbon receptor (Ahr) regulates the differentiation and function of CD4+ T cells; however, its cell-intrinsic role in CD8+ T cells remains elusive. Herein we show that Ahr acts as a promoter of resident memory CD8+ T cell (TRM) differentiation and function. Genetic ablation of Ahr in mouse CD8+ T cells leads to increased CD127-KLRG1+ short-lived effector cells and CD44+CD62L+ T central memory cells but reduced granzyme-B-producing CD69+CD103+ TRM cells. Genome-wide analyses reveal that Ahr suppresses the circulating while promoting the resident memory core gene program. A tumor resident polyfunctional CD8+ T cell population, revealed by single-cell RNA-seq, is diminished upon Ahr deletion, compromising anti-tumor immunity. Human intestinal intraepithelial CD8+ T cells also highly express AHR that regulates in vitro TRM differentiation and granzyme B production. Collectively, these data suggest that Ahr is an important cell-intrinsic factor for CD8+ T cell immunity.
Collapse
Affiliation(s)
- Joseph W Dean
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Eric Y Helm
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Zheng Fu
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Lifeng Xiong
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Na Sun
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Kristen N Oliff
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Marcus Muehlbauer
- Division of Gastroenterology, Hepatology and Nutrition, College of Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Dorina Avram
- Department of Immunology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Liang Zhou
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA.
| |
Collapse
|
32
|
Ortiz NR, Guy N, Garcia YA, Sivils JC, Galigniana MD, Cox MB. Functions of the Hsp90-Binding FKBP Immunophilins. Subcell Biochem 2023; 101:41-80. [PMID: 36520303 DOI: 10.1007/978-3-031-14740-1_2] [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] [Indexed: 12/23/2022]
Abstract
The Hsp90 chaperone is known to interact with a diverse array of client proteins. However, in every case examined, Hsp90 is also accompanied by a single or several co-chaperone proteins. One class of co-chaperone contains a tetratricopeptide repeat (TPR) domain that targets the co-chaperone to the C-terminal region of Hsp90. Within this class are Hsp90-binding peptidylprolyl isomerases, most of which belong to the FK506-binding protein (FKBP) family. Despite the common association of FKBP co-chaperones with Hsp90, it is abundantly clear that the client protein influences, and is often influenced by, the particular FKBP bound to Hsp90. Examples include Xap2 in aryl hydrocarbon receptor complexes and FKBP52 in steroid receptor complexes. In this chapter, we discuss the known functional roles played by FKBP co-chaperones and, where possible, relate distinctive functions to structural differences between FKBP members.
Collapse
Affiliation(s)
- Nina R Ortiz
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, USA
| | - Naihsuan Guy
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, USA
| | - Yenni A Garcia
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, USA
| | - Jeffrey C Sivils
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, USA
| | - Mario D Galigniana
- Departamento de Química Biológica/IQUIBICEN, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Biología y Medicina Experimental/CONICET, Buenos Aires, Argentina
| | - Marc B Cox
- Border Biomedical Research Center and Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, USA.
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX, USA.
| |
Collapse
|
33
|
Abudahab S, Price ET, Dozmorov MG, Deshpande LS, McClay JL. The Aryl Hydrocarbon Receptor, Epigenetics and the Aging Process. J Nutr Health Aging 2023; 27:291-300. [PMID: 37170437 PMCID: PMC10947811 DOI: 10.1007/s12603-023-1908-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-dependent transcription factor, classically associated with the regulation of xenobiotic metabolism in response to environmental toxins. In recent years, transgenic rodent models have implicated AhR in aging and longevity. Moreover, several AhR ligands, such as resveratrol and quercetin, are compounds proven to extend the lifespan of model organisms. In this paper, we first review AhR biology with a focus on aging and highlight several AhR ligands with potential anti-aging properties. We outline how AhR-driven expression of xenobiotic metabolism genes into old age may be a key mechanism through which moderate induction of AhR elicits positive benefits on longevity and healthspan. Furthermore, via integration of publicly available datasets, we show that liver-specific AhR target genes are enriched among genes subject to epigenetic aging. Changes to epigenetic states can profoundly affect transcription factor binding and are a hallmark of the aging process. We suggest that the interplay between AhR and epigenetic aging should be the subject of future research and outline several key gaps in the current literature. Finally, we recommend that a broad range of non-toxic AhR ligands should be investigated for their potential to promote healthspan and longevity.
Collapse
Affiliation(s)
- S Abudahab
- Sara Abudahab, Smith Building, 410 North 12th Street, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298-0533, USA.
| | | | | | | | | |
Collapse
|
34
|
Riaz F, Pan F, Wei P. Aryl hydrocarbon receptor: The master regulator of immune responses in allergic diseases. Front Immunol 2022; 13:1057555. [PMID: 36601108 PMCID: PMC9806217 DOI: 10.3389/fimmu.2022.1057555] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR) is a widely studied ligand-activated cytosolic transcriptional factor that has been associated with the initiation and progression of various diseases, including autoimmune diseases, cancers, metabolic syndromes, and allergies. Generally, AhR responds and binds to environmental toxins/ligands, dietary ligands, and allergens to regulate toxicological, biological, cellular responses. In a canonical signaling manner, activation of AhR is responsible for the increase in cytochrome P450 enzymes which help individuals to degrade and metabolize these environmental toxins and ligands. However, canonical signaling cannot be applied to all the effects mediated by AhR. Recent findings indicate that activation of AhR signaling also interacts with some non-canonical factors like Kruppel-like-factor-6 (KLF6) or estrogen-receptor-alpha (Erα) to affect the expression of downstream genes. Meanwhile, enormous research has been conducted to evaluate the effect of AhR signaling on innate and adaptive immunity. It has been shown that AhR exerts numerous effects on mast cells, B cells, macrophages, antigen-presenting cells (APCs), Th1/Th2 cell balance, Th17, and regulatory T cells, thus, playing a significant role in allergens-induced diseases. This review discussed how AhR mediates immune responses in allergic diseases. Meanwhile, we believe that understanding the role of AhR in immune responses will enhance our knowledge of AhR-mediated immune regulation in allergic diseases. Also, it will help researchers to understand the role of AhR in regulating immune responses in autoimmune diseases, cancers, metabolic syndromes, and infectious diseases.
Collapse
Affiliation(s)
- Farooq Riaz
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen, China
| | - Fan Pan
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen, China,*Correspondence: Ping Wei, ; Fan Pan,
| | - Ping Wei
- Department of Otolaryngology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China,*Correspondence: Ping Wei, ; Fan Pan,
| |
Collapse
|
35
|
Wang J, Lu P, Xie W. Atypical functions of xenobiotic receptors in lipid and glucose metabolism. MEDICAL REVIEW (2021) 2022; 2:611-624. [PMID: 36785576 PMCID: PMC9912049 DOI: 10.1515/mr-2022-0032] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/08/2022] [Indexed: 12/02/2022]
Abstract
Xenobiotic receptors are traditionally defined as xenobiotic chemical-sensing receptors, the activation of which transcriptionally regulates the expression of enzymes and transporters involved in the metabolism and disposition of xenobiotics. Emerging evidence suggests that "xenobiotic receptors" also have diverse endobiotic functions, including their effects on lipid metabolism and energy metabolism. Dyslipidemia is a major risk factor for cardiovascular disease, diabetes, obesity, metabolic syndrome, stroke, nonalcoholic fatty liver disease (NAFLD), and nonalcoholic steatohepatitis (NASH). Understanding the molecular mechanism by which transcriptional factors, including the xenobiotic receptors, regulate lipid homeostasis will help to develop preventive and therapeutic approaches. This review describes recent advances in our understanding the atypical roles of three xenobiotic receptors: aryl hydrocarbon receptor (AhR), pregnane X receptor (PXR), and constitutive androstane receptor (CAR), in metabolic disorders, with a particular focus on their effects on lipid and glucose metabolism. Collectively, the literatures suggest the potential values of AhR, PXR and CAR as therapeutic targets for the treatment of NAFLD, NASH, obesity and diabetes, and cardiovascular diseases.
Collapse
Affiliation(s)
- Jingyuan Wang
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peipei Lu
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Wen Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
36
|
From Nucleus to Organs: Insights of Aryl Hydrocarbon Receptor Molecular Mechanisms. Int J Mol Sci 2022; 23:ijms232314919. [PMID: 36499247 PMCID: PMC9738205 DOI: 10.3390/ijms232314919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022] Open
Abstract
The aryl hydrocarbon receptor (AHR) is a markedly established regulator of a plethora of cellular and molecular processes. Its initial role in the detoxification of xenobiotic compounds has been partially overshadowed by its involvement in homeostatic and organ physiology processes. In fact, the discovery of its ability to bind specific target regulatory sequences has allowed for the understanding of how AHR modulates such processes. Thereby, AHR presents functions in transcriptional regulation, chromatin architecture modifications and participation in different key signaling pathways. Interestingly, such fields of influence end up affecting organ and tissue homeostasis, including regenerative response both to endogenous and exogenous stimuli. Therefore, from classical spheres such as canonical transcriptional regulation in embryonic development, cell migration, differentiation or tumor progression to modern approaches in epigenetics, senescence, immune system or microbiome, this review covers all aspects derived from the balance between regulation/deregulation of AHR and its physio-pathological consequences.
Collapse
|
37
|
Patil NY, Rus I, Downing E, Mandala A, Friedman JE, Joshi AD. Cinnabarinic Acid Provides Hepatoprotection Against Nonalcoholic Fatty Liver Disease. J Pharmacol Exp Ther 2022; 383:32-43. [PMID: 35933113 PMCID: PMC9513857 DOI: 10.1124/jpet.122.001301] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 07/12/2022] [Indexed: 11/22/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a chronic condition in which excess lipids accumulate in the liver and can lead to a range of progressive liver disorders including non-alcoholic steatohepatitis, liver cirrhosis, and hepatocellular carcinoma. While lifestyle and diet modifications have proven to be effective as NAFLD treatments, they are not sustainable in the long-term, and currently no pharmacological therapies are approved to treat NAFLD. Our previous studies demonstrated that cinnabarinic acid (CA), a novel endogenous Aryl hydrocarbon Receptor (AhR) agonist, activates the AhR target gene, Stanniocalcin 2, and confers cytoprotection against a plethora of ER/oxidative stressors. In this study, the hepatoprotective and anti-steatotic properties of CA were examined against free fatty-acid-induced in vitro and high-fat-diet fed in vivo NAFLD models. The results demonstrated that CA treatment significantly lowered weight gain and attenuated hepatic lipotoxicity both before and after the established fatty liver, thereby protecting against steatosis, inflammation, and liver injury. CA mitigated intracellular free fatty acid uptake concomitant with the downregulation of CD36/fatty acid translocase. Genes involved in fatty acid and triglyceride synthesis were also downregulated in response to CA treatment. Additionally, suppressing AhR and Stc2 expression using RNA interference in vitro verified that the hepatoprotective effects of CA were absolutely dependent on both AhR and its target, Stc2. Collectively, our results demonstrate that the endogenous AhR agonist, CA, confers hepatoprotection against NAFLD by regulating hepatic fatty acid uptake and lipogenesis. SIGNIFICANCE STATEMENT: In this study using in vitro and in vivo models, we demonstrate that cinnabarinic acid (CA), an endogenous AhR agonist, provides protection against non-alcoholic fatty liver disease. CA bestows cytoprotection against steatosis and liver injury by controlling expression of several key genes associated with lipid metabolism pathways, limiting the hepatic lipid uptake, and controlling liver inflammation. Moreover, CA-induced hepatoprotection is absolutely dependent on AhR and Stc2 expression.
Collapse
Affiliation(s)
- Nikhil Y Patil
- Department of Pharmaceutical Sciences (N.Y.P., I.R., E.D., A.D.J.) and Harold Hamm Diabetes Center (A.M., J.E.F., A.D.J.), University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Iulia Rus
- Department of Pharmaceutical Sciences (N.Y.P., I.R., E.D., A.D.J.) and Harold Hamm Diabetes Center (A.M., J.E.F., A.D.J.), University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Emma Downing
- Department of Pharmaceutical Sciences (N.Y.P., I.R., E.D., A.D.J.) and Harold Hamm Diabetes Center (A.M., J.E.F., A.D.J.), University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Ashok Mandala
- Department of Pharmaceutical Sciences (N.Y.P., I.R., E.D., A.D.J.) and Harold Hamm Diabetes Center (A.M., J.E.F., A.D.J.), University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Jacob E Friedman
- Department of Pharmaceutical Sciences (N.Y.P., I.R., E.D., A.D.J.) and Harold Hamm Diabetes Center (A.M., J.E.F., A.D.J.), University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Aditya D Joshi
- Department of Pharmaceutical Sciences (N.Y.P., I.R., E.D., A.D.J.) and Harold Hamm Diabetes Center (A.M., J.E.F., A.D.J.), University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
38
|
Liver regeneration after partial hepatectomy is improved in the absence of aryl hydrocarbon receptor. Sci Rep 2022; 12:15446. [PMID: 36104446 PMCID: PMC9474532 DOI: 10.1038/s41598-022-19733-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 09/02/2022] [Indexed: 11/19/2022] Open
Abstract
The liver is among the few organs having the ability to self-regenerate in response to a severe damage compromising its functionality. The Aryl hydrocarbon receptor (Ahr) is a transcription factor relevant for the detoxification of xenobiotics but also largely important for liver development and homeostasis. Hence, liver cell differentiation is developmentally modulated by Ahr through the controlled expression of pluripotency and stemness-inducing genes. Here, 2/3 partial hepatectomy (PH) was used as a clinically relevant approach to induce liver regeneration in Ahr-expressing (Ahr+/+) and Ahr-null (Ahr−/−) mice. Ahr expression and activity were early induced after 2/3 PH to be gradually downmodulated latter during regeneration. Ahr−/− mice triggered liver regeneration much faster than AhR+/+ animals, although both reached full regeneration at the latest times. At initial stages after PHx, earlier regenerating Ahr−/− livers had upregulation of cell proliferation markers and increased activation of signalling pathways related to stemness such as Hippo-YAP and Wnt/β-catenin, concomitantly with the induction of pro-inflammatory cytokines TNFa, IL6 and p65. These phenotypes, together with the improved metabolic adaptation of Ahr−/− mice after PHx and their induced sustained cell proliferation, could likely result from the expansion of undifferentiated stem cells residing in the liver expressing OCT4, SOX2, KLF4 and NANOG. We propose that Ahr needs to be induced early during regeneration to fine-tune liver regrowth to physiological values. Since Ahr deficiency did not result in liver overgrowth, its transient pharmacological inhibition could serve to improve liver regeneration in hepatectomized and transplanted patients and in those exposed to damaging liver toxins and carcinogens.
Collapse
|
39
|
Salminen A. Aryl hydrocarbon receptor (AhR) reveals evidence of antagonistic pleiotropy in the regulation of the aging process. Cell Mol Life Sci 2022; 79:489. [PMID: 35987825 PMCID: PMC9392714 DOI: 10.1007/s00018-022-04520-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/14/2022] [Accepted: 08/08/2022] [Indexed: 11/24/2022]
Abstract
The antagonistic pleiotropy hypothesis is a well-known evolutionary theory to explain the aging process. It proposes that while a particular gene may possess beneficial effects during development, it can exert deleterious properties in the aging process. The aryl hydrocarbon receptor (AhR) has a significant role during embryogenesis, but later in life, it promotes several age-related degenerative processes. For instance, AhR factor (i) controls the pluripotency of stem cells and the stemness of cancer stem cells, (ii) it enhances the differentiation of embryonal stem cells, especially AhR signaling modulates the differentiation of hematopoietic stem cells and progenitor cells, (iii) it also stimulates the differentiation of immunosuppressive Tregs, Bregs, and M2 macrophages, and finally, (iv) AhR signaling participates in the differentiation of many peripheral tissues. On the other hand, AhR signaling is involved in many processes promoting cellular senescence and pathological processes, e.g., osteoporosis, vascular dysfunction, and the age-related remodeling of the immune system. Moreover, it inhibits autophagy and aggravates extracellular matrix degeneration. AhR signaling also stimulates oxidative stress, promotes excessive sphingolipid synthesis, and disturbs energy metabolism by catabolizing NAD+ degradation. The antagonistic pleiotropy of AhR signaling is based on the complex and diverse connections with major signaling pathways in a context-dependent manner. The major regulatory steps include, (i) a specific ligand-dependent activation, (ii) modulation of both genetic and non-genetic responses, (iii) a competition and crosstalk with several transcription factors, such as ARNT, HIF-1α, E2F1, and NF-κB, and (iv) the epigenetic regulation of target genes with binding partners. Thus, not only mTOR signaling but also the AhR factor demonstrates antagonistic pleiotropy in the regulation of the aging process.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland.
| |
Collapse
|
40
|
Salminen A. Mutual antagonism between aryl hydrocarbon receptor and hypoxia-inducible factor-1α (AhR/HIF-1α) signaling: Impact on the aging process. Cell Signal 2022; 99:110445. [PMID: 35988806 DOI: 10.1016/j.cellsig.2022.110445] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/27/2022] [Accepted: 08/16/2022] [Indexed: 11/26/2022]
Abstract
The ambient oxygen level, many environmental toxins, and the rays of ultraviolet light (UV) provide a significant risk for the maintenance of organismal homeostasis. The aryl hydrocarbon receptors (AhR) represent a complex sensor system not only for environmental toxins and UV radiation but also for many endogenous ligands, e.g., L-tryptophan metabolites. The AhR signaling system is evolutionarily conserved and AhR homologs existed as many as 600 million years ago. The ancient atmosphere demanded the evolution of an oxygen-sensing system, i.e., hypoxia-inducible transcription factors (HIF) and their prolyl hydroxylase regulators (PHD). Given that both signaling systems have important roles in embryogenesis, it seems that they have been involved in the evolution of multicellular organisms. The evolutionary origin of the aging process is unknown although it is most likely associated with the evolution of multicellularity. Intriguingly, there is compelling evidence that while HIF-1α signaling extends the lifespan, that of AhR promotes many age-related degenerative processes, e.g., it increases oxidative stress, inhibits autophagy, promotes cellular senescence, and aggravates extracellular matrix degeneration. In contrast, HIF-1α signaling stimulates autophagy, inhibits cellular senescence, and enhances cell proliferation. Interestingly, there is a clear antagonism between the AhR and HIF-1α signaling pathways. For instance, (i) AhR and HIF-1α factors heterodimerize with the same factor, ARNT/HIF-1β, leading to their competition for DNA-binding, (ii) AhR and HIF-1α signaling exert antagonistic effects on autophagy, and (iii) co-chaperone p23 exhibits specific functions in the signaling of AhR and HIF-1α factors. One might speculate that it is the competition between the AhR and HIF-1α signaling pathways that is a driving force in the aging process.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| |
Collapse
|
41
|
Yang CE, Wang YN, Hua MR, Miao H, Zhao YY, Cao G. Aryl hydrocarbon receptor: From pathogenesis to therapeutic targets in aging-related tissue fibrosis. Ageing Res Rev 2022; 79:101662. [PMID: 35688331 DOI: 10.1016/j.arr.2022.101662] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 04/22/2022] [Accepted: 06/02/2022] [Indexed: 11/27/2022]
Abstract
Aging promotes chronic inflammation, which contributes to fibrosis and decreases organ function. Fibrosis, the excessive synthesis and deposition of extracellular matrix components, is the main cause of most chronic diseases including aging-related organ failure. Organ fibrosis in the heart, liver, and kidneys is the final manifestation of many chronic diseases. The aryl hydrocarbon receptor (AHR) is a cytoplasmic receptor and highly conserved transcription factor that is activated by a variety of small-molecule ligands to affect a wide array of tissue homeostasis functions. In recent years, mounting evidence has revealed that AHR plays an important role in multi-organ fibrosis initiation, progression, and therapy. In this review, we summarise the relationship between AHR and the pathogenesis of aging-related tissue fibrosis, and further discuss how AHR modulates tissue fibrosis by regulating transforming growth factor-β signalling, immune response, and mitochondrial function, which may offer novel targets for the prevention and treatment of this condition.
Collapse
Affiliation(s)
- Chang-E Yang
- Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi 710069, China
| | - Yan-Ni Wang
- Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi 710069, China
| | - Meng-Ru Hua
- Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi 710069, China
| | - Hua Miao
- Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi 710069, China.
| | - Ying-Yong Zhao
- Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi 710069, China.
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang 310053, China.
| |
Collapse
|
42
|
Phillips JL, Buermeyer AB, Nguyen BD, Loehr C, Kolluri SK. Loss of the aryl hydrocarbon receptor increases tumorigenesis in p53-deficient mice. Toxicol Appl Pharmacol 2022; 454:116191. [PMID: 35926564 DOI: 10.1016/j.taap.2022.116191] [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: 05/20/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 11/29/2022]
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that regulates cell fate via activation of a diverse set of genes. There are conflicting reports describing the role of AhR in cancer. AhR-knockout mice do not develop tumors spontaneously, yet the AhR can act as a tumor suppressor in certain contexts. Loss of tumor suppression by p53 is common in human cancer. To investigate AhR function in the absence of p53, we generated mice lacking both AhR and p53. Mice deficient for AhR and p53 had shortened lifespan, increased tumorigenesis, and an altered tumor spectrum relative to control mice lacking only p53. In addition, knockout of both AhR and p53 resulted in reduced embryonic survival and neonatal fitness. We also examined the consequences of loss of AhR in p53-heterozygous mice and observed a significantly reduced lifespan and enhanced tumor burden. These findings reveal an important role for the AhR as a tumor suppressor in the absence of p53 signaling and support the development of anti-cancer therapeutics that would promote the tumor suppressive actions of the AhR.
Collapse
Affiliation(s)
- Jessica L Phillips
- Cancer Research Laboratory, Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, OR 97331, United States of America
| | - Andrew B Buermeyer
- Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, OR 897331, United States of America; The Pacific Northwest Center for Translational Environmental Health Research, Oregon State University, Corvallis, OR 97331, USA
| | - Bach D Nguyen
- Cancer Research Laboratory, Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, OR 97331, United States of America
| | - Christiane Loehr
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR 897331, United States of America; The Pacific Northwest Center for Translational Environmental Health Research, Oregon State University, Corvallis, OR 97331, USA
| | - Siva K Kolluri
- Cancer Research Laboratory, Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, OR 97331, United States of America; The Pacific Northwest Center for Translational Environmental Health Research, Oregon State University, Corvallis, OR 97331, USA.
| |
Collapse
|
43
|
Yu HX, Feng Z, Lin W, Yang K, Liu RQ, Li JQ, Liu XY, Pei M, Yang HT. Ongoing Clinical Trials in Aging-Related Tissue Fibrosis and New Findings Related to AhR Pathways. Aging Dis 2022; 13:732-752. [PMID: 35656117 PMCID: PMC9116921 DOI: 10.14336/ad.2021.1105] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 11/05/2021] [Indexed: 11/06/2022] Open
Abstract
Fibrosis is a pathological manifestation of wound healing that replaces dead/damaged tissue with collagen-rich scar tissue to maintain homeostasis, and complications from fibrosis contribute to nearly half of all deaths in the industrialized world. Ageing is closely associated with a progressive decline in organ function, and the prevalence of tissue fibrosis dramatically increases with age. Despite the heavy clinical and economic burden of organ fibrosis as the population ages, to date, there is a paucity of therapeutic strategies that are specifically designed to slow fibrosis. Aryl hydrocarbon receptor (AhR) is an environment-sensing transcription factor that exacerbates aging phenotypes in different tissues that has been brought back into the spotlight again with economic development since AhR could interact with persistent organic pollutants derived from incomplete waste combustion. In addition, gut microbiota dysbiosis plays a pivotal role in the pathogenesis of numerous diseases, and microbiota-associated tryptophan metabolites are dedicated contributors to fibrogenesis by acting as AhR ligands. Therefore, a better understanding of the effects of tryptophan metabolites on fibrosis modulation through AhR may facilitate the exploitation of new therapeutic avenues for patients with organ fibrosis. In this review, we primarily focus on how tryptophan-derived metabolites are involved in renal fibrosis, idiopathic pulmonary fibrosis, hepatic fibrosis and cardiac fibrosis. Moreover, a series of ongoing clinical trials are highlighted.
Collapse
Affiliation(s)
- Hang-Xing Yu
- 1Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,2National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Zhe Feng
- 3Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Wei Lin
- 1Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,2National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Kang Yang
- 4Kidney Disease Treatment Center, The first affiliated hospital of Henan university of CM, Zhengzhou, Henan, China
| | - Rui-Qi Liu
- 1Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,2National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Jia-Qi Li
- 1Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,2National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xin-Yue Liu
- 1Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,2National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Ming Pei
- 1Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,2National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Hong-Tao Yang
- 1Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,2National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
44
|
Nacarino-Palma A, Rico-Leo EM, Campisi J, Ramanathan A, González-Rico FJ, Rejano-Gordillo CM, Ordiales-Talavero A, Merino JM, Fernández-Salguero PM. Aryl hydrocarbon receptor blocks aging-induced senescence in the liver and fibroblast cells. Aging (Albany NY) 2022; 14:4281-4304. [PMID: 35619220 PMCID: PMC9186759 DOI: 10.18632/aging.204103] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 03/06/2022] [Indexed: 01/10/2023]
Abstract
Aging impairs organismal homeostasis leading to multiple pathologies. Yet, the mechanisms and molecular intermediates involved are largely unknown. Here, we report that aged aryl hydrocarbon receptor-null mice (AhR-/-) had exacerbated cellular senescence and more liver progenitor cells. Senescence-associated markers β-galactosidase (SA-β-Gal), p16Ink4a and p21Cip1 and genes encoding senescence-associated secretory phenotype (SASP) factors TNF and IL1 were overexpressed in aged AhR-/- livers. Chromatin immunoprecipitation showed that AhR binding to those gene promoters repressed their expression, thus adjusting physiological levels in AhR+/+ livers. MCP-2, MMP12 and FGF secreted by senescent cells were overproduced in aged AhR-null livers. Supporting the relationship between senescence and stemness, liver progenitor cells were overrepresented in AhR-/- mice, probably contributing to increased hepatocarcinoma burden. These AhR roles are not liver-specific since adult and embryonic AhR-null fibroblasts underwent senescence in culture, overexpressing SA-β-Gal, p16Ink4a and p21Cip1. Notably, depletion of senescent cells with the senolytic agent navitoclax restored expression of senescent markers in AhR-/- fibroblasts, whereas senescence induction by palbociclib induced an AhR-null-like phenotype in AhR+/+ fibroblasts. AhR levels were downregulated by senescence in mouse lungs but restored upon depletion of p16Ink4a-expressing senescent cells. Thus, AhR restricts age-induced senescence associated to a differentiated phenotype eventually inducing resistance to liver tumorigenesis.
Collapse
Affiliation(s)
- Ana Nacarino-Palma
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, Universidad de Extremadura, Badajoz 06071, Spain.,Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), Badajoz 06071, Spain
| | - Eva M Rico-Leo
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, Universidad de Extremadura, Badajoz 06071, Spain.,Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), Badajoz 06071, Spain
| | - Judith Campisi
- Buck Institute for Research on Aging, Novato, CA 94945, USA.,Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | | | - Francisco J González-Rico
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, Universidad de Extremadura, Badajoz 06071, Spain.,Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), Badajoz 06071, Spain
| | - Claudia M Rejano-Gordillo
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, Universidad de Extremadura, Badajoz 06071, Spain.,Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), Badajoz 06071, Spain
| | - Ana Ordiales-Talavero
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, Universidad de Extremadura, Badajoz 06071, Spain.,Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), Badajoz 06071, Spain
| | - Jaime M Merino
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, Universidad de Extremadura, Badajoz 06071, Spain.,Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), Badajoz 06071, Spain
| | - Pedro M Fernández-Salguero
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, Universidad de Extremadura, Badajoz 06071, Spain.,Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), Badajoz 06071, Spain
| |
Collapse
|
45
|
Damasceno LEA, Cebinelli GCM, Fernandes MF, Nascimento DC, Públio GA, Vinolo MAR, Oliveira SC, Sparwasser T, Cunha TM, Cunha FQ, Alves-Filho JC. STING is an intrinsic checkpoint inhibitor that restrains the T H17 cell pathogenic program. Cell Rep 2022; 39:110838. [PMID: 35613599 PMCID: PMC9188824 DOI: 10.1016/j.celrep.2022.110838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 03/02/2022] [Accepted: 04/28/2022] [Indexed: 12/01/2022] Open
Abstract
External and intrinsic factors regulate the transcriptional profile of T helper 17 (TH17) cells, thereby affecting their pathogenic potential and revealing their context-dependent plasticity. The stimulator of interferon genes (STING), a component of the intracellular DNA-sensing pathway, triggers immune responses but remains largely unexplored in T cells. Here, we describe an intrinsic role of STING in limiting the TH17 cell pathogenic program. We demonstrate that non-pathogenic TH17 cells express higher levels of STING than those activated under pathogenic conditions. Activation of STING induces interleukin-10 (IL-10) production in TH17 cells, decreasing IL-17A and IL-23R expression in a type I interferon (IFN)-independent manner. Mechanistically, STING-induced IL-10 production partially requires aryl hydrocarbon receptor (AhR) signaling, while the decrease of IL-17A expression occurs due to a reduction of Rorγt transcriptional activity. Our findings reveal a regulatory function of STING in the TH17 cell activation program, proposing it as a valuable target to limit TH17-cell-mediated inflammation. TH17 cells display a spectrum of pathogenic states depending on environmental and intrinsic cues. Damasceno et al. demonstrate that STING activation induces a non-pathogenic TH17 profile. Mechanistically, STING impairs Rorγt-mediated Il17a transcription, thereby reducing IL-17A production. Besides that, STING activation promotes IL-10 expression through AhR signaling pathway.
Collapse
Affiliation(s)
- Luis Eduardo Alves Damasceno
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil
| | - Guilherme Cesar Martelossi Cebinelli
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil
| | - Mariane Font Fernandes
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP 13083-862, Brazil
| | - Daniele Carvalho Nascimento
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil
| | - Gabriel Azevedo Públio
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil
| | - Marco Aurélio Ramirez Vinolo
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP 13083-862, Brazil
| | - Sergio Costa Oliveira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Tim Sparwasser
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University, Mainz 55131, Germany
| | - Thiago Mattar Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil
| | - Fernando Queiroz Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil
| | - José Carlos Alves-Filho
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil.
| |
Collapse
|
46
|
Larigot L, Bui LC, de Bouvier M, Pierre O, Pinon G, Fiocca J, Ozeir M, Tourette C, Ottolenghi C, Imbeaud S, Pontoizeau C, Blaise BJ, Chevallier A, Tomkiewicz C, Legrand B, Elena-Herrmann B, Néri C, Brinkmann V, Nioche P, Barouki R, Ventura N, Dairou J, Coumoul X. Identification of Modulators of the C. elegans Aryl Hydrocarbon Receptor and Characterization of Transcriptomic and Metabolic AhR-1 Profiles. Antioxidants (Basel) 2022; 11:antiox11051030. [PMID: 35624894 PMCID: PMC9137885 DOI: 10.3390/antiox11051030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 05/06/2022] [Accepted: 05/18/2022] [Indexed: 02/02/2023] Open
Abstract
The Aryl hydrocarbon Receptor (AhR) is a xenobiotic sensor in vertebrates, regulating the metabolism of its own ligands. However, no ligand has been identified to date for any AhR in invertebrates. In C. elegans, the AhR ortholog, AHR-1, displays physiological functions. Therefore, we compared the transcriptomic and metabolic profiles of worms expressing AHR-1 or not and investigated the putative panel of chemical AHR-1 modulators. The metabolomic profiling indicated a role for AHR-1 in amino acids, carbohydrates, and fatty acids metabolism. The transcriptional profiling in neurons expressing AHR-1, identified 95 down-regulated genes and 76 up-regulated genes associated with neuronal and metabolic functions in the nervous system. A gene reporter system allowed us to identify several AHR-1 modulators including bacterial, dietary, or environmental compounds. These results shed new light on the biological functions of AHR-1 in C. elegans and perspectives on the evolution of the AhR functions across species.
Collapse
Affiliation(s)
- Lucie Larigot
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, Université Paris Cité, 75006 Paris, France; (L.L.); (L.-C.B.); (M.d.B.); (O.P.); (G.P.); (J.F.); (M.O.); (C.O.); (A.C.); (C.T.); (B.L.); (P.N.); (R.B.)
- CNRS UMR 8601, Metabolism, Pharmacochemistry and Neurochemistry, Université Paris Cité, 75006 Paris, France
| | - Linh-Chi Bui
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, Université Paris Cité, 75006 Paris, France; (L.L.); (L.-C.B.); (M.d.B.); (O.P.); (G.P.); (J.F.); (M.O.); (C.O.); (A.C.); (C.T.); (B.L.); (P.N.); (R.B.)
- Unité de biologie fonctionnelle et adaptative, UMR 8251, CNRS, Université Paris Cité, 75013 Paris, France
| | - Marine de Bouvier
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, Université Paris Cité, 75006 Paris, France; (L.L.); (L.-C.B.); (M.d.B.); (O.P.); (G.P.); (J.F.); (M.O.); (C.O.); (A.C.); (C.T.); (B.L.); (P.N.); (R.B.)
| | - Ophélie Pierre
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, Université Paris Cité, 75006 Paris, France; (L.L.); (L.-C.B.); (M.d.B.); (O.P.); (G.P.); (J.F.); (M.O.); (C.O.); (A.C.); (C.T.); (B.L.); (P.N.); (R.B.)
- Laboratoire Interactions Epithéliums-Neurones (LIEN), Université de Brest, EA4685, 29200 Brest, France
| | - Grégory Pinon
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, Université Paris Cité, 75006 Paris, France; (L.L.); (L.-C.B.); (M.d.B.); (O.P.); (G.P.); (J.F.); (M.O.); (C.O.); (A.C.); (C.T.); (B.L.); (P.N.); (R.B.)
- Structural and Molecular Analysis Platform, Biomedtech Facilities, Université Paris Cité, 75006 Paris, France
| | - Justine Fiocca
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, Université Paris Cité, 75006 Paris, France; (L.L.); (L.-C.B.); (M.d.B.); (O.P.); (G.P.); (J.F.); (M.O.); (C.O.); (A.C.); (C.T.); (B.L.); (P.N.); (R.B.)
- Structural and Molecular Analysis Platform, Biomedtech Facilities, Université Paris Cité, 75006 Paris, France
| | - Mohammad Ozeir
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, Université Paris Cité, 75006 Paris, France; (L.L.); (L.-C.B.); (M.d.B.); (O.P.); (G.P.); (J.F.); (M.O.); (C.O.); (A.C.); (C.T.); (B.L.); (P.N.); (R.B.)
- Structural and Molecular Analysis Platform, Biomedtech Facilities, Université Paris Cité, 75006 Paris, France
| | - Cendrine Tourette
- Centre Paul Broca, INSERM U894 Neuronal Cell Biology & Pathology & EA Université Paris Cité, 75014 Paris, France;
| | - Chris Ottolenghi
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, Université Paris Cité, 75006 Paris, France; (L.L.); (L.-C.B.); (M.d.B.); (O.P.); (G.P.); (J.F.); (M.O.); (C.O.); (A.C.); (C.T.); (B.L.); (P.N.); (R.B.)
- AP-HP, Hôpital Necker-Enfants Malades, Service de Biochimie Métabolique, 75015 Paris, France;
| | - Sandrine Imbeaud
- Gif/Orsay DNA MicroArray Platform, 91190 Gif sur Yvette, France;
| | - Clément Pontoizeau
- AP-HP, Hôpital Necker-Enfants Malades, Service de Biochimie Métabolique, 75015 Paris, France;
- Centre de Résonance Magnétique Nucléaire à Très Hauts Champs, Univ. Lyon, CNRS, UCBL, ENS Lyon, 69100 Villeurbanne, France; (B.J.B.); (B.E.-H.)
| | - Benjamin J. Blaise
- Centre de Résonance Magnétique Nucléaire à Très Hauts Champs, Univ. Lyon, CNRS, UCBL, ENS Lyon, 69100 Villeurbanne, France; (B.J.B.); (B.E.-H.)
| | - Aline Chevallier
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, Université Paris Cité, 75006 Paris, France; (L.L.); (L.-C.B.); (M.d.B.); (O.P.); (G.P.); (J.F.); (M.O.); (C.O.); (A.C.); (C.T.); (B.L.); (P.N.); (R.B.)
| | - Céline Tomkiewicz
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, Université Paris Cité, 75006 Paris, France; (L.L.); (L.-C.B.); (M.d.B.); (O.P.); (G.P.); (J.F.); (M.O.); (C.O.); (A.C.); (C.T.); (B.L.); (P.N.); (R.B.)
| | - Béatrice Legrand
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, Université Paris Cité, 75006 Paris, France; (L.L.); (L.-C.B.); (M.d.B.); (O.P.); (G.P.); (J.F.); (M.O.); (C.O.); (A.C.); (C.T.); (B.L.); (P.N.); (R.B.)
| | - Bénédicte Elena-Herrmann
- Centre de Résonance Magnétique Nucléaire à Très Hauts Champs, Univ. Lyon, CNRS, UCBL, ENS Lyon, 69100 Villeurbanne, France; (B.J.B.); (B.E.-H.)
- Institute for Advanced Biosciences, Univ. Grenoble Alpes, CNRS, INSERM, 38000 Grenoble, France
| | - Christian Néri
- CNRS UMR 8256, Inserm ERL U1164, Sorbonne Université, 75005 Paris, France;
| | - Vanessa Brinkmann
- Institute of Clinical Chemistry and Laboratory Diagnostic, Medical Faculty, Heinrich Heine University, Düsseldorf, Moorenstr 5, 40225 Düsseldorf, Germany; (V.B.); (N.V.)
- Leibniz Institute for Environmental Medicine (IUF), Auf’m Hennekamp 50, 40225 Düsseldorf, Germany
| | - Pierre Nioche
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, Université Paris Cité, 75006 Paris, France; (L.L.); (L.-C.B.); (M.d.B.); (O.P.); (G.P.); (J.F.); (M.O.); (C.O.); (A.C.); (C.T.); (B.L.); (P.N.); (R.B.)
- Structural and Molecular Analysis Platform, Biomedtech Facilities, Université Paris Cité, 75006 Paris, France
| | - Robert Barouki
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, Université Paris Cité, 75006 Paris, France; (L.L.); (L.-C.B.); (M.d.B.); (O.P.); (G.P.); (J.F.); (M.O.); (C.O.); (A.C.); (C.T.); (B.L.); (P.N.); (R.B.)
- Assistance Publique-Hôpitaux de Paris, Hôpital Necker, 75015 Paris, France
| | - Natascia Ventura
- Institute of Clinical Chemistry and Laboratory Diagnostic, Medical Faculty, Heinrich Heine University, Düsseldorf, Moorenstr 5, 40225 Düsseldorf, Germany; (V.B.); (N.V.)
- Leibniz Institute for Environmental Medicine (IUF), Auf’m Hennekamp 50, 40225 Düsseldorf, Germany
| | - Julien Dairou
- CNRS UMR 8601, Metabolism, Pharmacochemistry and Neurochemistry, Université Paris Cité, 75006 Paris, France
- Correspondence: (J.D.); (X.C.); Tel.: +33-1-42-86-91-21 (J.D.); +33-1-42-86-33-59 (X.C.)
| | - Xavier Coumoul
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, Université Paris Cité, 75006 Paris, France; (L.L.); (L.-C.B.); (M.d.B.); (O.P.); (G.P.); (J.F.); (M.O.); (C.O.); (A.C.); (C.T.); (B.L.); (P.N.); (R.B.)
- Correspondence: (J.D.); (X.C.); Tel.: +33-1-42-86-91-21 (J.D.); +33-1-42-86-33-59 (X.C.)
| |
Collapse
|
47
|
Aryl Hydrocarbon Receptors: Evidence of Therapeutic Targets in Chronic Inflammatory Skin Diseases. Biomedicines 2022; 10:biomedicines10051087. [PMID: 35625824 PMCID: PMC9139118 DOI: 10.3390/biomedicines10051087] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/01/2022] [Accepted: 05/02/2022] [Indexed: 02/04/2023] Open
Abstract
The aryl hydrocarbon receptor (AhR), a ligand-dependent transcription factor, is important for xenobiotic metabolism and binds to various endogenous and exogenous ligands present in the skin. AhR is known to be associated with diseases in various organs; however, its functions in chronic inflammatory skin diseases, such as atopic dermatitis (AD) and psoriasis (PS), have recently been elucidated. Here, we discuss the molecular mechanisms of AhR related to chronic inflammatory skin diseases, such as AD and PS, and the mechanisms of action of AhR on the skin immune system. The importance of AhR molecular biological pathways, clinical features in animal models, and AhR ligands in skin diseases need to be investigated. In conclusion, the therapeutic effects of AhR ligands are demonstrated based on the relationship between AhR and skin diseases. Nevertheless, further studies are required to elucidate the detailed roles of AhR in chronic inflammatory skin diseases.
Collapse
|
48
|
Zhou J, Blevins LK, Crawford RB, Kaminski NE. Role of Programmed Cell Death Protein-1 and Lymphocyte Specific Protein Tyrosine Kinase in the Aryl Hydrocarbon Receptor- Mediated Impairment of the IgM Response in Human CD5 + Innate-Like B Cells. Front Immunol 2022; 13:884203. [PMID: 35558082 PMCID: PMC9088000 DOI: 10.3389/fimmu.2022.884203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 03/28/2022] [Indexed: 11/24/2022] Open
Abstract
Innate-like B cells (ILBs) are a heterogeneous population B cells which participate in innate and adaptive immune responses. This diverse subset of B cells is characterized by the expression of CD5 and has been shown to secrete high levels of immunoglobulin M (IgM) in the absence of infection or vaccination. Further, CD5+ ILBs have been shown to express high basal levels of lymphocyte specific protein tyrosine kinase (LCK) and programmed cell death protein-1 (PD-1), which are particularly sensitive to stimulation by interferon gamma (IFNγ). Previous studies have demonstrated that activation of the aryl hydrocarbon receptor (AHR), a cytosolic ligand-activated transcription factor, results in suppressed IgM responses and is dependent on LCK. A recent study showed that CD5+ ILBs are particularly sensitive to AHR activation as evidenced by a significant suppression of the IgM response compared to CD5- B cells, which were refractory. Therefore, the objective of this study was to further investigate the role of LCK and PD-1 signaling in AHR-mediated suppression of CD5+ ILBs. In addition, studies were conducted to establish whether IFNγ alters the levels of LCK and PD-1 in CD5+ ILBs. We found that AHR activation led to a significant upregulation of total LCK and PD-1 proteins in CD5+ ILBs, which correlated with suppression of IgM. Interestingly, treatment with recombinant IFNγ reduced LCK protein levels and reversed AHR-mediated IgM suppression in CD5+ ILBs in a similar manner as LCK inhibitors. Collectively, these results support a critical role for LCK and PD-1 in AHR-mediated suppression of the IgM response in human CD5+ ILBs.
Collapse
Affiliation(s)
- Jiajun Zhou
- Department of Microbiology & Molecular Genetics, Michigan State University, East Lansing, MI, United States
- Institute of Integrative Toxicology, Michigan State University, East Lansing, MI, United States
| | - Lance K. Blevins
- Institute of Integrative Toxicology, Michigan State University, East Lansing, MI, United States
| | - Robert B. Crawford
- Institute of Integrative Toxicology, Michigan State University, East Lansing, MI, United States
| | - Norbert E. Kaminski
- Institute of Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI, United States
- Center for Research on Ingredient Safety, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
49
|
Rosmarinus officinalis L. Leaf Extracts and Their Metabolites Inhibit the Aryl Hydrocarbon Receptor (AhR) Activation In Vitro and in Human Keratinocytes: Potential Impact on Inflammatory Skin Diseases and Skin Cancer. Molecules 2022; 27:molecules27082499. [PMID: 35458697 PMCID: PMC9029298 DOI: 10.3390/molecules27082499] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/01/2022] [Accepted: 04/10/2022] [Indexed: 12/02/2022] Open
Abstract
Aryl hydrocarbon receptor (AhR) activation by environmental agents and microbial metabolites is potentially implicated in a series of skin diseases. Hence, it would be very important to identify natural compounds that could inhibit the AhR activation by ligands of microbial origin as 6-formylindolo[3,2-b]carbazole (FICZ), indirubin (IND) and pityriazepin (PZ) or the prototype ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Five different dry Rosmarinus officinalis L. extracts (ROEs) were assayed for their activities as antagonists of AhR ligand binding with guinea pig cytosol in the presence of [3H]TCDD. The methanolic ROE was further assayed towards CYP1A1 mRNA induction using RT-PCR in human keratinocytes against TCDD, FICZ, PZ, and IND. The isolated metabolites, carnosic acid, carnosol, 7-O-methyl-epi-rosmanol, 4′,7-O-dimethylapigenin, and betulinic acid, were assayed for their agonist and antagonist activity in the presence and absence of TCDD using the gel retardation assay (GRA). All assayed ROE extracts showed similar dose-dependent activities with almost complete inhibition of AhR activation by TCDD at 100 ppm. The methanol ROE at 10 ppm showed 99%, 50%, 90%, and 85% inhibition against TCDD, FICZ, IND, and PZ, respectively, in human keratinocytes. Most assayed metabolites exhibited dose-dependent antagonist activity. ROEs inhibit AhR activation by TCDD and by the Malassezia metabolites FICZ, PZ, and IND. Hence, ROE could be useful for the prevention or treatment of skin diseases mediated by activation of AhR.
Collapse
|
50
|
Rejano-Gordillo C, Ordiales-Talavero A, Nacarino-Palma A, Merino JM, González-Rico FJ, Fernández-Salguero PM. Aryl Hydrocarbon Receptor: From Homeostasis to Tumor Progression. Front Cell Dev Biol 2022; 10:884004. [PMID: 35465323 PMCID: PMC9022225 DOI: 10.3389/fcell.2022.884004] [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: 02/25/2022] [Accepted: 03/15/2022] [Indexed: 12/19/2022] Open
Abstract
Transcription factor aryl hydrocarbon receptor (AHR) has emerged as one of the main regulators involved both in different homeostatic cell functions and tumor progression. Being a member of the family of basic-helix-loop-helix (bHLH) transcriptional regulators, this intracellular receptor has become a key member in differentiation, pluripotency, chromatin dynamics and cell reprogramming processes, with plenty of new targets identified in the last decade. Besides this role in tissue homeostasis, one enthralling feature of AHR is its capacity of acting as an oncogene or tumor suppressor depending on the specific organ, tissue and cell type. Together with its well-known modulation of cell adhesion and migration in a cell-type specific manner in epithelial-mesenchymal transition (EMT), this duality has also contributed to the arise of its clinical interest, highlighting a new potential as therapeutic tool, diagnosis and prognosis marker. Therefore, a deregulation of AHR-controlled pathways may have a causal role in contributing to physiological and homeostatic failures, tumor progression and dissemination. With that firmly in mind, this review will address the remarkable capability of AHR to exert a different function influenced by the phenotype of the target cell and its potential consequences.
Collapse
Affiliation(s)
- Claudia Rejano-Gordillo
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Spain
| | - Ana Ordiales-Talavero
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Spain
| | - Ana Nacarino-Palma
- Chronic Diseases Research Centre (CEDOC), Rua Do Instituto Bacteriológico, Lisboa, Portugal
| | - Jaime M. Merino
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Spain
| | - Francisco J. González-Rico
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Spain
- *Correspondence: Francisco J. González-Rico, ; Pedro M. Fernández-Salguero,
| | - Pedro M. Fernández-Salguero
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Spain
- *Correspondence: Francisco J. González-Rico, ; Pedro M. Fernández-Salguero,
| |
Collapse
|