1
|
Palrasu M, Marudamuthu A, Kakar K, Hamida H, Thada S, Gupta R, Wilson K, Carter T, Zhong Y, Saxena A, Yang X, Singh N, Busbee PB, Li J, Garcia-Buitrago M, Nagarkatti P, Nagarkatti M. AhR-Dependent Induction of β-Defensin 1 in Colonic Epithelial Cells Regulates Cross-Talk between Gut Microbiota and Immune Response Leading to Attenuation of Colitis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2416324. [PMID: 40410944 DOI: 10.1002/advs.202416324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 04/16/2025] [Indexed: 05/26/2025]
Abstract
The aryl hydrocarbon receptor (AhR) acts as a critical signaling hub that connects immune cells, food and environmental cues, and microbiota to regulate intestinal homeostasis. In the current study, the role of AhR in the regulation of an antimicrobial peptide, β-defensin1 (BD-1) is investigated to control colitis. Human patients with ulcerative colitis (UC) and Crohn's disease (CD), and mice with three different models of colitis, express a significant decrease in the expression of BD-1 in colonic epithelial cells (CECs). Dietary and environmental AhR ligands induce the expression of BD-1 in CECs through the activation of two dioxin-responsive elements (DREs) expressed on its promoter. AhR ligands attenuate colitis in wild-type (WT) mice while inducing BD-1. However, AhR ligands fail to induce BD-1 and protect mice from colitis when there is an intestinal epithelial cell (IEC)-specific deletion of AhR. Blocking BD1 in vivo using antibodies prevents the ability of AhR ligands to ameliorate colitis, restore dysbiosis, and attenuate colonic inflammation. The current study identifies a novel pathway involving dietary, environmental, and endogenous AhR ligands to induce the antimicrobial peptide BD-1 in IECs, which in turn, plays a critical role in the regulation of intestinal homeostasis.
Collapse
Affiliation(s)
- Manikandan Palrasu
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, 29208, USA
| | - Amarnath Marudamuthu
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, 29208, USA
| | - Khadija Kakar
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, 29208, USA
| | - Hamida Hamida
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, 29208, USA
| | - Shruthi Thada
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, 29208, USA
| | - Rohan Gupta
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, 29208, USA
| | - Kiesha Wilson
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, 29208, USA
| | - Taylor Carter
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, 29208, USA
| | - Yin Zhong
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, 29208, USA
| | - Archana Saxena
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, 29208, USA
| | - Xiaoming Yang
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, 29208, USA
| | - Narendra Singh
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, 29208, USA
| | - Philip Brandon Busbee
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, 29208, USA
| | - Jie Li
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, 29208, USA
| | - Monica Garcia-Buitrago
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Prakash Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, 29208, USA
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, 29208, USA
| |
Collapse
|
2
|
Wei Q, Jiang H, Zeng J, Xu J, Zhang H, Xiao E, Lu Q, Huang G. Quercetin protected the gut barrier in ulcerative colitis by activating aryl hydrocarbon receptor. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156633. [PMID: 40088746 DOI: 10.1016/j.phymed.2025.156633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 02/25/2025] [Accepted: 03/08/2025] [Indexed: 03/17/2025]
Abstract
BACKGROUND Ulcerative colitis (UC) is characterized by abdominal pain and bloody diarrhoea and restoring the gut barrier is the core goal of UC treatment. Activation of aryl hydrocarbon receptor (Ahr) was reported to effectively alleviate symptoms and repair the gut barrier damage. Neutrophil extracellular traps (NETs) have been recognized as potential targets in the treatment of UC. Ahr activation has been found to be capable of upregulating Nqo1, thereby reducing the production of reactive oxygen species (ROS), which is important in the formation of NETs. Quercetin (QUE), which is derived from natural plants and herbs used in traditional Chinese medicine (TCM), is able to strengthen gut barrier function by activating Ahr. PURPOSE The aim of this study is to investigate how QUE suppresses NETs in UC and activates Ahr in neutrophils. METHODS In this study, the dextran sulfate sodium (DSS)-induced UC model was used. Histopathological assessments were performed in the paraffin slides of tissues after H&E, PAS, Masson and alcian blue staining. The concentration of cytokines was also detected using cytometric beads array kits. Based on the transcriptomic analysis of colon tissues, western blot (WB) analysis, immunohistochemistry (IHC) assays and immunofluorescence (IF) assays were conducted to validate the significantly regulated genes and pathways. In vitro, the binding of quercetin to Ahr was calculated by molecular dynamic simulations (MDS) and biolayer interferometry (BLI) analysis. Primary neutrophils isolated from mice were cocultured with LPS or PMA with or without quercetin. The regulated genes were detected using WB, real-time quantitative PCR, enzyme-linked immunosorbent assay (ELISA) and IF analysis. The agonists and antagonist of Ahr were used as the control. RESULTS After the administration of quercetin, colon inflammation and gut barrier disruption was significantly prevented through inhibiting the NF-κB pathway and upregulating the expression of Ahr/Arnt and Nqo1. The transcriptomic analysis and IHC assays showed that inflammation and NETs were greatly decreased by QUE treatment. In vitro, quercetin inhibited LPS-induced inflammatory responses through NF-κB pathway. Furthermore, MDS and BLI analysis revealed that QUE is an agonist of AHR. QUE activated Ahr translocation and reduced ROS production via regulation of Arnt and Nqo1. CONCLUSION This study proved that quercetin greatly improved gut barrier function in the DSS-induced colitis model by regulating NET formation and that quercetin was able to activate Ahr and upregulate Arnt in neutrophils to regulate NET formation.
Collapse
Affiliation(s)
- Qiuzhu Wei
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Haixu Jiang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China; School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Jia Zeng
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Jie Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Honglin Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Enfan Xiao
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Qingyi Lu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Guangrui Huang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China.
| |
Collapse
|
3
|
Hu Y, Chen LL, Ye Z, Li LZ, Qian HZ, Wu MQ, Wang J, Qin KH, Ye QB. Indigo naturalis as a potential drug in the treatment of ulcerative colitis: a comprehensive review of current evidence. PHARMACEUTICAL BIOLOGY 2024; 62:818-832. [PMID: 39475104 PMCID: PMC11533244 DOI: 10.1080/13880209.2024.2415652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 08/25/2024] [Accepted: 10/03/2024] [Indexed: 11/06/2024]
Abstract
CONTEXT Ulcerative colitis (UC) is an intractable inflammatory bowel disease that threatens the health of patients. The limited availability of therapeutic strategies makes it imperative to explore more efficient and safer drugs. Indigo naturalis (IN) is a traditional Chinese medicine that possesses many pharmacological activities, including anti-inflammatory, antioxidant, and immunomodulatory activities. The treatment potential of IN for UC has been proven by numerous preclinical and clinical studies in recent years. OBJECTIVE This article provides a comprehensive review of the utility and potential of IN in the treatment of UC. METHODS 'Indigo naturalis' 'Qing dai' 'Qingdai' 'Ulcerative colitis' and 'UC' are used as the keywords, and the relevant literature is collected from online databases (Elsevier, PubMed, and Web of Science). RESULTS AND CONCLUSION Indirubin, indigo, isatin, tryptanthrin, and β-sitosterol are considered the key components in the treatment of UC with IN. Both preclinical and clinical studies support the efficacy of IN for UC, especially in severe UC or in those who do not respond to or have poor efficacy with existing therapies. The mechanisms of IN for UC are associated with the aryl hydrocarbon receptor pathway activation, immune regulation, oxidative stress inhibition, and intestinal microbial modulation. However, the clinical use of IN has the risks of adverse events such as pulmonary hypertension, which suggests the necessity for its rational application. As a potential therapeutic agent for UC that is currently receiving more attention, the clinical value of IN has been initially demonstrated and warrants further evaluation.
Collapse
Affiliation(s)
- Yu Hu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liu-lin Chen
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhen Ye
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lin-zhen Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huan-zhu Qian
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ming-quan Wu
- Department of Pharmacy, Sichuan Provincial Orthopedic Hospital, Chengdu, China
| | - Juan Wang
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kai-hua Qin
- Health Preservation and Rehabilitation College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiao-bo Ye
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
4
|
Huang Y, Liu J, Liang D. Comprehensive analysis reveals key genes and environmental toxin exposures underlying treatment response in ulcerative colitis based on in-silico analysis and Mendelian randomization. Aging (Albany NY) 2023; 15:14141-14171. [PMID: 38059894 PMCID: PMC10756092 DOI: 10.18632/aging.205294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 11/03/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND UC is increasingly prevalent worldwide and represents a significant global disease burden. Although medical therapeutics are employed, they often fall short of being optimal, leaving patients struggling with treatment non-responsiveness and many related complications. MATERIALS AND METHODS The study utilized gene microarray data and clinical information from GEO. Gene enrichment and differential expression analyses were conducted using Metascape and Limma, respectively. Lasso Regression Algorithm was constructed using glmnet and heat maps were generated using pheatmap. ROC curves were used to assess diagnostic parameter capability, while XSum was employed to screen for small-molecule drugs exacerbating UC. Molecular docking was carried out using Autodock Vina. The study also performed Mendelian randomization analysis based on TwoSampleMR and used CTD to investigate the relationship between exposure to environmental chemical toxicants and UC therapy responsiveness. RESULTS Six genes (ELL2, DAPP1, SAMD9L, CD38, IGSF6, and LYN) were found to be significantly overexpressed in UC patient samples that did not respond to multiple therapies. Lasso analysis identified ELL2 and DAPP1 as key genes influencing UC treatment response. Both genes accurately predicted intestinal inflammation in UC and impacted the immunological infiltration status. Clofibrate showed therapeutic potential for UC by binding to ELL2 and DAPP1 proteins. The study also reviews environmental toxins and drug exposures that could impact UC progression. CONCLUSIONS We used microarray technology to identify DAPP1 and ELL2 as key genes that impact UC treatment response and inflammatory progression. Clofibrate was identified as a promising UC treatment. Our review also highlights the impact of environmental toxins on UC treatment response, providing valuable insights for personalized clinical management.
Collapse
Affiliation(s)
- Yizhou Huang
- Department of Gastroenterology, The PLA Navy Anqing Hospital, Anqing 246000, Anhui Province, China
| | - Jie Liu
- Department of Gastroenterology, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, Anhui Province, China
| | - Dingbao Liang
- Department of Gastroenterology, The PLA Navy Anqing Hospital, Anqing 246000, Anhui Province, China
| |
Collapse
|
5
|
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
|
6
|
Lai Y, Wu X, Chao E, Bloomstein JD, Wei G, Hwang ST, Shi Z. Impact of Gut Bacterial Metabolites on Psoriasis and Psoriatic Arthritis: Current Status and Future Perspectives. J Invest Dermatol 2023; 143:1657-1666. [PMID: 37422760 DOI: 10.1016/j.jid.2023.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 05/04/2023] [Accepted: 05/17/2023] [Indexed: 07/10/2023]
Abstract
There is growing evidence that supports a role of gut dysbiosis in the pathogenesis of psoriasis (Pso). Thus, probiotic supplementation and fecal microbiota transplantation may serve as promising preventive and therapeutic strategies for patients with Pso. One of the basic mechanisms through which the gut microbiota interacts with the host is through bacteria-derived metabolites, usually intermediate or end products produced by microbial metabolism. In this study, we provide an up-to-date review of the most recent literature on microbial-derived metabolites and highlight their roles in the immune system, with a special focus on Pso and one of its most common comorbidities, psoriatic arthritis.
Collapse
Affiliation(s)
- Yuhsien Lai
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xuesong Wu
- Department of Dermatology, University of California, Davis, Sacramento, California, USA
| | - Ellen Chao
- Department of Dermatology, University of California, Davis, Sacramento, California, USA
| | | | - Grace Wei
- Department of Dermatology, University of California, Davis, Sacramento, California, USA
| | - Sam T Hwang
- Department of Dermatology, University of California, Davis, Sacramento, California, USA
| | - Zhenrui Shi
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
7
|
Huang RG, Li XB, Wang YY, Wu H, Li KD, Jin X, Du YJ, Wang H, Qian FY, Li BZ. Endocrine-disrupting chemicals and autoimmune diseases. ENVIRONMENTAL RESEARCH 2023; 231:116222. [PMID: 37224951 DOI: 10.1016/j.envres.2023.116222] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 04/10/2023] [Accepted: 05/21/2023] [Indexed: 05/26/2023]
Abstract
Endocrine-disrupting chemicals (EDCs) widely exist in people's production and life which have great potential to damage human and animal health. Over the past few decades, growing attention has been paid to the impact of EDCs on human health, as well as immune system. So far, researchers have proved that EDCs (such as bisphenol A (BPA), phthalate, tetrachlorodibenzodioxin (TCDD), etc.) affect human immune function and promotes the occurrence and development of autoimmune diseases (ADs). Therefore, in order to better understand how EDCs affect ADs, we summarized the current knowledge about the impact of EDCs on ADs, and elaborated the potential mechanism of the impact of EDCs on ADs in this review.
Collapse
Affiliation(s)
- Rong-Gui Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Xian-Bao Li
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Yi-Yu Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Hong Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Kai-Di Li
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Xue Jin
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Yu-Jie Du
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Hua Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | | | - Bao-Zhu Li
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China.
| |
Collapse
|
8
|
Dong F, Murray IA, Annalora A, Coslo D, Desai D, Gowda K, Yang J, Wang D, Koo I, Hao F, Amin SG, Patterson AD, Marcus C, Perdew GH. Complex chemical signals dictate Ah receptor activation through the gut-lung axis. FASEB J 2023; 37:e23010. [PMID: 37272852 PMCID: PMC10264151 DOI: 10.1096/fj.202300703r] [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: 04/10/2023] [Revised: 05/16/2023] [Accepted: 05/19/2023] [Indexed: 06/06/2023]
Abstract
The aryl hydrocarbon receptor (AHR) mediates intestinal barrier homeostasis. Many AHR ligands are also CYP1A1/1B1 substrates, which can result in rapid clearance within the intestinal tract, limiting systemic exposure and subsequent AHR activation. This led us to the hypothesis that there are dietary substrates of CYP1A1/1B1 that functionally increase the half-life of potent AHR ligands. We examined the potential of urolithin A (UroA), a gut bacterial metabolite of ellagitannins, as a CYP1A1/1B1 substrate to enhance AHR activity in vivo. UroA is a competitive substrate for CYP1A1/1B1 in an in vitro competition assay. A broccoli-containing diet promotes the gastric formation of the potent hydrophobic AHR ligand and CYP1A1/1B1 substrate, 5,11-dihydroindolo[3,2-b]carbazole (ICZ). In mice, dietary exposure to UroA in a 10% broccoli diet led to a coordinated increase in duodenal, cardiac, and pulmonary AHR activity, but no increase in activity in the liver. Thus, CYP1A1 dietary competitive substrates can lead to enhanced systemic AHR ligand distribution from the gut, likely through the lymphatic system, increasing AHR activation in key barrier tissues. Finally, this report will lead to a reassessment of the dynamics of distribution of other hydrophobic chemicals present in the diet.
Collapse
Affiliation(s)
- Fangcong Dong
- Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802, USA
| | - Iain A. Murray
- Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802, USA
| | - Andrew Annalora
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, USA
| | - Denise Coslo
- Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802, USA
| | - Dhimant Desai
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, 17033 USA
| | - Krishne Gowda
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, 17033 USA
| | - Jian Yang
- Department of Biomedical Engineering, Materials Research Institute, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Dingbowen Wang
- Department of Biomedical Engineering, Materials Research Institute, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Imhoi Koo
- Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802, USA
| | - Fuhua Hao
- Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802, USA
| | - Shantu G. Amin
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, 17033 USA
| | - Andrew D. Patterson
- Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802, USA
| | - Craig Marcus
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, USA
| | - Gary H. Perdew
- Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
9
|
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
|
10
|
Dong F, Murray IA, Annalora A, Coslo D, Desai D, Gowda K, Yang J, Wang D, Koo I, Hao F, Amin SG, Patterson AD, Marcus C, Perdew GH. Complex chemical signals dictate Ah receptor activation through the gut-lung axis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.22.529529. [PMID: 36865156 PMCID: PMC9980078 DOI: 10.1101/2023.02.22.529529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
The aryl hydrocarbon receptor (AHR) mediates intestinal barrier homeostasis. Many AHR ligands are also CYP1A1/1B1 substrates, which can result in the rapid clearance within the intestinal tract, limiting AHR activation. This led us to the hypothesis that there are dietary substrates of CYP1A1/1B1 that increase the half-life of potent AHR ligands. We examined the potential of urolithin A (UroA) as a CYP1A1/1B1 substrate to enhance AHR activity in vivo. UroA is a competitive substrate for CYP1A1/1B1 in an in vitro competition assay. A broccoli-containing diet promotes the gastric formation of the potent hydrophobic AHR ligand and CYP1A1/1B1 substrate, 5,11-dihydroindolo[3,2-b]carbazole (ICZ). Dietary exposure to UroA in a broccoli diet led to a coordinated increase in duodenal, cardiac, and pulmonary AHR activity, but no increase in activity in liver. Thus, CYP1A1 dietary competitive substrates can lead to intestinal "escape", likely through the lymphatic system, increasing AHR activation in key barrier tissues.
Collapse
|
11
|
Forde B, Yao L, Shaha R, Murphy S, Lunjani N, O'Mahony L. Immunomodulation by foods and microbes: Unravelling the molecular tango. Allergy 2022; 77:3513-3526. [PMID: 35892227 PMCID: PMC10087875 DOI: 10.1111/all.15455] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/15/2022] [Accepted: 07/23/2022] [Indexed: 01/28/2023]
Abstract
Metabolic health and immune function are intimately connected via diet and the microbiota. Nearly 90% of all immune cells in the body are associated with the gastrointestinal tract and these immune cells are continuously exposed to a wide range of microbes and microbial-derived compounds, with important systemic ramifications. Microbial dysbiosis has consistently been observed in patients with atopic dermatitis, food allergy and asthma and the molecular mechanisms linking changes in microbial populations with disease risk and disease endotypes are being intensively investigated. The discovery of novel bacterial metabolites that impact immune function is at the forefront of host-microbe research. Co-evolution of microbial communities within their hosts has resulted in intertwined metabolic pathways that affect physiological and pathological processes. However, recent dietary and lifestyle changes are thought to negatively influence interactions between microbes and their host. This review provides an overview of some of the critical metabolite-receptor interactions that have been recently described, which may underpin the immunomodulatory effects of the microbiota, and are of relevance for allergy, asthma and infectious diseases.
Collapse
Affiliation(s)
- Brian Forde
- APC Microbiome Ireland, UCC, Cork, Ireland.,School of Microbiology, UCC, Cork, Ireland
| | - Lu Yao
- APC Microbiome Ireland, UCC, Cork, Ireland.,School of Microbiology, UCC, Cork, Ireland
| | - Rupin Shaha
- APC Microbiome Ireland, UCC, Cork, Ireland.,School of Microbiology, UCC, Cork, Ireland
| | | | - Nonhlanhla Lunjani
- APC Microbiome Ireland, UCC, Cork, Ireland.,University of Cape Town, Cape Town, South Africa
| | - Liam O'Mahony
- APC Microbiome Ireland, UCC, Cork, Ireland.,School of Microbiology, UCC, Cork, Ireland.,Department of Medicine, UCC, Cork, Ireland
| |
Collapse
|
12
|
Cannon AR, Shim EH, Kuprys PV, Choudhry MA. IL-22 and Lactobacillus delbrueckii mitigate alcohol-induced exacerbation of DSS-induced colitis. J Leukoc Biol 2022; 112:1471-1484. [PMID: 35916052 PMCID: PMC9701151 DOI: 10.1002/jlb.4a0122-068r] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 06/29/2022] [Indexed: 01/04/2023] Open
Abstract
Ulcerative colitis (UC) is characterized by cycles of active disease flare and inactive disease remission. During UC remission, IL-22 is up-regulated, acting as a hallmark of entrance into UC remission. Recently, we found that in our mouse model of binge alcohol and dextran sodium sulfate (DSS)-induced colitis, alcohol increases severity of UC pathology. In this study, we assessed not only whether alcohol influenced IL-22 expression and thereby perpetuates UC, but also whether recombinant IL-22 (rIL-22) or treatment with a probiotic could alleviate exacerbated symptoms of UC. Levels of large intestine IL-22 were significantly decreased ∼6.9-fold in DSS ethanol compared with DSS vehicle. Examination of lamina propria (LP) cells in the large intestine revealed IL-22+ γδ T cells in DSS vehicle-treated mice were significantly increased, while IL-22+ γδ T cells in DSS ethanol mice were unable to mount this IL-22 response. We administered rIL-22 and found it restored weight loss of DSS ethanol-treated mice. Colonic shortening and increased Enterobacteriaceae were also attenuated. Administration of Lactobacillus delbrueckii attenuated weight loss (p < 0.01), colon length (p < 0.001), mitigated increases in Enterobacteriaceae, increased levels of IL-22, and increased levels of p-STAT3 back to that of DSS vehicle group in DSS ethanol mice. In contrast, sole administration of L. delbrueckii supernatant was not sufficient to reduce UC exacerbation following alcohol. Our findings suggest L. delbrueckii contributes to repair mechanisms by increasing levels of IL-22, resulting in phosphorylation of STAT3, thus attenuating the alcohol-induced increases in intestinal damage after colitis.
Collapse
Affiliation(s)
- Abigail R. Cannon
- Alcohol Research Program, Burn and Shock Trauma Research Institute, Department of Surgery, Loyola University Chicago Health Sciences Campus, Maywood, IL 60153, USA
- Integrative Cell Biology Program, Loyola University Chicago Health Sciences Campus, Maywood, IL 60153, USA
- Department of Surgery, Loyola University Chicago Health Sciences Campus, Maywood, IL 60153, USA
| | - Esther H. Shim
- Alcohol Research Program, Burn and Shock Trauma Research Institute, Department of Surgery, Loyola University Chicago Health Sciences Campus, Maywood, IL 60153, USA
| | - Paulius V. Kuprys
- Alcohol Research Program, Burn and Shock Trauma Research Institute, Department of Surgery, Loyola University Chicago Health Sciences Campus, Maywood, IL 60153, USA
| | - Mashkoor A. Choudhry
- Alcohol Research Program, Burn and Shock Trauma Research Institute, Department of Surgery, Loyola University Chicago Health Sciences Campus, Maywood, IL 60153, USA
- Integrative Cell Biology Program, Loyola University Chicago Health Sciences Campus, Maywood, IL 60153, USA
- Department of Surgery, Loyola University Chicago Health Sciences Campus, Maywood, IL 60153, USA
- Department of Microbiology and Immunology, Loyola University Chicago Health Sciences Campus, Maywood, IL 60153, USA
| |
Collapse
|
13
|
Tryptamine, a Microbial Metabolite in Fermented Rice Bran Suppressed Lipopolysaccharide-Induced Inflammation in a Murine Macrophage Model. Int J Mol Sci 2022; 23:ijms231911209. [PMID: 36232510 PMCID: PMC9570467 DOI: 10.3390/ijms231911209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/15/2022] [Accepted: 09/19/2022] [Indexed: 11/16/2022] Open
Abstract
Fermentation is thought to alter the composition and bioavailability of bioactive compounds in rice bran. However, how this process affects the anti-inflammatory effects of rice bran and the bioactive compounds that might participate in this function is yet to be elucidated. This study aimed to isolate bioactive compounds in fermented rice bran that play a key role in its anti-inflammatory function. The fermented rice bran was fractionated using a succession of solvent and solid-phase extractions. The fermented rice bran fractions were then applied to lipopolysaccharide (LPS)-activated murine macrophages to evaluate their anti-inflammatory activity. The hot water fractions (FRBA), 50% ethanol fractions (FRBB), and n-hexane fractions (FRBC) were all shown to be able to suppress the pro-inflammatory cytokine expression from LPS-stimulated RAW 264.7 cells. Subsequent fractions from the hot water fraction (FRBF and FRBE) were also able to reduce the inflammatory response of these cells to LPS. Further investigation revealed that tryptamine, a bacterial metabolite of tryptophan, was abundantly present in these extracts. These results indicate that tryptamine may play an important role in the anti-inflammatory effects of fermented rice bran. Furthermore, the anti-inflammatory effects of FRBE and tryptamine may depend on the activity of the aryl hydrocarbon receptor.
Collapse
|
14
|
Tu P, Chi L, Bian X, Gao B, Ru H, Lu K. A Black Raspberry-Rich Diet Protects From Dextran Sulfate Sodium-Induced Intestinal Inflammation and Host Metabolic Perturbation in Association With Increased Aryl Hydrocarbon Receptor Ligands in the Gut Microbiota of Mice. Front Nutr 2022; 9:842298. [PMID: 35734371 PMCID: PMC9208328 DOI: 10.3389/fnut.2022.842298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 04/21/2022] [Indexed: 11/13/2022] Open
Abstract
Dietary modulation of the gut microbiota recently received considerable attention, and ligand activation of aryl hydrocarbon receptor (AHR) plays a pivotal role in intestinal immunity. Importantly, black raspberry (BRB, Rubus occidentalis) is associated with a variety of beneficial health effects. We aim to investigate effects of a BRB-rich diet on dextran sulfate sodium (DSS)-induced intestinal inflammation and to determine whether its consequent anti-inflammatory effects are relevant to modulation of the gut microbiota, especially its production of AHR ligands. A mouse model of DSS-induced intestinal inflammation was used in the present study. C57BL/6J mice were fed either AIN-76A or BRB diet. Composition and functions of the gut microbiota were assessed by 16S rRNA sequencing and comparative metagenome analysis. Metabolic profiles of host and the gut microbiome were assessed by serum and fecal metabolomic profiling and identification. BRB diet was found to ameliorate DSS-induced intestinal inflammation and host metabolic perturbation. BRB diet also protected from DSS-induced perturbation in diversity and composition in the gut microbiota. BRB diet promoted AHR ligand production by the gut microbiota, as revealed by increased levels of fecal AHR activity in addition to increased levels of two known AHR ligands, hemin and biliverdin. Accordingly, enrichment of bacterial genes and pathways responsible for production of hemin and biliverdin were found, specific gut bacteria that are highly correlated with abundances of hemin and biliverdin were also identified. BRB dietary intervention ameliorated intestinal inflammation in mice in association with promotion of AHR ligand production by the gut microbiota.
Collapse
Affiliation(s)
- Pengcheng Tu
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Liang Chi
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Xiaoming Bian
- Department of Environmental Health Sciences, University of Georgia, Athens, GA, United States
| | - Bei Gao
- Department of Environmental Health Sciences, University of Georgia, Athens, GA, United States
| | - Hongyu Ru
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kun Lu
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
15
|
Reduced Colonic Mucosal Injury in 2,3,7,8-Tetrachlorodibenzo- p-Dioxin Poly ADP-Ribose Polymerase (TIPARP/PARP7)-Deficient Mice. Int J Mol Sci 2022; 23:ijms23020920. [PMID: 35055106 PMCID: PMC8779828 DOI: 10.3390/ijms23020920] [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: 12/16/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 01/27/2023] Open
Abstract
Poly-ADP-ribose polymerases (PARPs) are important regulators of the immune system, including TCDD-inducible poly-ADP-ribose polymerase (TIPARP), also known as poly-ADP-ribose polymerase 7 (PARP7). PARP7 negatively regulates aryl hydrocarbon receptor (AHR) and type I interferon (IFN-I) signaling, both of which have been implicated in intestinal homeostasis and immunity. Since the loss of PARP7 expression increases AHR and IFN-I signaling, we used a murine dextran sulfate sodium (DSS)-induced colitis model to investigate the effect of PARP7 loss on DSS-induced intestinal inflammation. DSS-exposed Parp7−/− mice had less body weight loss, lower disease index scores, and reduced expression of several inflammation genes, including interleukin IL-6, C-x-c motif chemokine ligand 1 (Cxcl1), and lipocalin-2, when compared with wild-type mice. However, no significant difference was observed between genotypes in the colonic expression of the AHR target gene cytochrome P450 1A1 (Cyp1a1). Moreover, no significant differences in microbial composition were observed between the genotypes. Our findings demonstrate that the absence of PARP7 protein results in an impaired immune response to colonic inflammation and suggests that PARP7 may participate in the recruitment of immune cells to the inflammation site, which may be due to its role in IFN-I signaling rather than AHR signaling.
Collapse
|
16
|
Oral benzo[a]pyrene administration attenuates dextran sulfate sodium-induced colitis in mice. Chem Biol Interact 2022; 353:109802. [PMID: 34998820 DOI: 10.1016/j.cbi.2022.109802] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/28/2021] [Accepted: 01/04/2022] [Indexed: 11/23/2022]
Abstract
Benzo[a]pyrene (BaP) is an environmental pollutant produced by combustion processes and is present in grilled foods as well as in tobacco smoke. BaP acts as an agonist for the aryl hydrocarbon receptor (AHR), and is metabolized by AHR-inducing enzymes. BaP metabolism can result in either detoxification or metabolic activation, the latter leads to an increased risk of disease, particularly lung cancer and cardiovascular disease, in a context-dependent manner. Although AHR activation has been thought to protect against inflammatory bowel disease, it remains unknown whether BaP exerts a protective or deleterious effect on colitis. In this study, we examined the effect of oral BaP administration on colitis induced by dextran sulfate sodium (DSS) in mice, an animal model of inflammatory bowel disease. BaP administration attenuated weight loss, shortening of the colon, disease activity index scores, and histological damage in DSS-induced colitis mice. BaP also suppressed colonic expression of inflammation-associated genes and plasma interleukin-6 secretion induced by DSS treatment. BaP-DNA adduct formation, a marker of BaP metabolic activation, was not enhanced in the colon after DSS treatment. Thus, oral BaP exerts an anti-inflammatory effect on DSS-induced colitis, without the toxicity associated with metabolic activation. The results provide insights into the disease-specific roles of BaP.
Collapse
|
17
|
Han H, Safe S, Jayaraman A, Chapkin RS. Diet-Host-Microbiota Interactions Shape Aryl Hydrocarbon Receptor Ligand Production to Modulate Intestinal Homeostasis. Annu Rev Nutr 2021; 41:455-478. [PMID: 34633858 PMCID: PMC8667662 DOI: 10.1146/annurev-nutr-043020-090050] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated basic-helix-loop-helix transcription factor that binds structurally diverse ligands and senses cues from environmental toxicants and physiologically relevant dietary/microbiota-derived ligands. The AhR is an ancient conserved protein and is widely expressed across different tissues in vertebrates and invertebrates. AhR signaling mediates a wide range of cellular functions in a ligand-, cell type-, species-, and context-specific manner. Dysregulation of AhR signaling is linked to many developmental defects and chronic diseases. In this review, we discuss the emerging role of AhR signaling in mediating bidirectional host-microbiome interactions. We also consider evidence showing the potential for the dietary/microbial enhancement ofhealth-promoting AhR ligands to improve clinical pathway management in the context of inflammatory bowel diseases and colon tumorigenesis.
Collapse
Affiliation(s)
- Huajun Han
- Program in Integrative Nutrition and Complex Diseases and Department of Nutrition, Texas A&M University, College Station, Texas 77843, USA;
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, Texas 77843, USA
| | - Stephen Safe
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, Texas 77843, USA
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843, USA
| | - Arul Jayaraman
- Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843, USA
| | - Robert S Chapkin
- Program in Integrative Nutrition and Complex Diseases and Department of Nutrition, Texas A&M University, College Station, Texas 77843, USA;
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, Texas 77843, USA
| |
Collapse
|
18
|
Pernomian L, Duarte-Silva M, de Barros Cardoso CR. The Aryl Hydrocarbon Receptor (AHR) as a Potential Target for the Control of Intestinal Inflammation: Insights from an Immune and Bacteria Sensor Receptor. Clin Rev Allergy Immunol 2021; 59:382-390. [PMID: 32279195 DOI: 10.1007/s12016-020-08789-3] [Citation(s) in RCA: 152] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The aryl hydrocarbon receptor (AHR) is widely expressed in immune and non-immune cells of the gut and its activation has been correlated to the outcome of inflammatory bowel diseases (IBD). In ulcerative colitis and Crohn's disease, there is an excessive chronic inflammation with massive accumulation of leukocytes in the gut, in an attempt to constrain the invasion of pathogenic microorganisms on the damaged organ. Accordingly, it is known that dietary components, xenobiotics, and some chemicals or metabolites can activate AHR and induce the modulation of inflammatory responses. In fact, the AHR triggering by specific ligands during inflammatory conditions results in decreased IFNγ, IL-6, IL-12, TNF, IL-7, and IL-17, along with reduced microbial translocation and fibrosis in the gut. Moreover, upon AHR activation, there are increased regulatory mechanisms such as IL-10, IL-22, prostaglandin E2, and Foxp3, besides the production of anti-microbial peptides and epithelial repair. Most interestingly, commensal bacteria or their metabolites may also activate this receptor, thus contributing to the restoration of gut normobiosis and homeostasis. In line with that, Lactobacillus reuteri, Lactobacillus bulgaricus, or microbial products such as tryptophan metabolites, indole-3-pyruvic acid, urolithin A, short-chain fatty acids, dihydroxyquinoline, and others may regulate the inflammation by mechanisms dependent on AHR activation. Hence, here we discussed the potential modulatory role of AHR on intestinal inflammation, focused on the reestablishment of homeostasis through the receptor triggering by microbial metabolites. Finally, the development of AHR-based therapies derived from bacteria products could represent an important future alternative for controlling IBD.
Collapse
Affiliation(s)
- Larissa Pernomian
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Murillo Duarte-Silva
- Department of Biochemistry and Immunology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Cristina Ribeiro de Barros Cardoso
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.
| |
Collapse
|
19
|
Grifka-Walk HM, Jenkins BR, Kominsky DJ. Amino Acid Trp: The Far Out Impacts of Host and Commensal Tryptophan Metabolism. Front Immunol 2021; 12:653208. [PMID: 34149693 PMCID: PMC8213022 DOI: 10.3389/fimmu.2021.653208] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/11/2021] [Indexed: 12/12/2022] Open
Abstract
Tryptophan (Trp) is an essential amino acid primarily derived from the diet for use by the host for protein synthesis. The intestinal tract is lined with cells, both host and microbial, that uptake and metabolize Trp to also generate important signaling molecules. Serotonin (5-HT), kynurenine and its downstream metabolites, and to a lesser extent other neurotransmitters are generated by the host to signal onto host receptors and elicit physiological effects. 5-HT production by neurons in the CNS regulates sleep, mood, and appetite; 5-HT production in the intestinal tract by enterochromaffin cells regulates gastric motility and inflammation in the periphery. Kynurenine can signal onto the aryl hydrocarbon receptor (AHR) to elicit pleiotropic responses from several cell types including epithelial and immune cells, or can be further metabolized into bioactive molecules to influence neurodegenerative disease. There is a remarkable amount of cross-talk with the microbiome with regard to tryptophan metabolites as well. The gut microbiome can regulate the production of host tryptophan metabolites and can use dietary or recycled trp to generate bioactive metabolites themselves. Trp derivatives like indole are able to signal onto xenobiotic receptors, including AHR, to elicit tolerogenic effects. Here, we review studies that demonstrate that tryptophan represents a key intra-kingdom signaling molecule.
Collapse
Affiliation(s)
| | | | - Douglas J. Kominsky
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| |
Collapse
|
20
|
Nolan LS, Mihi B, Agrawal P, Gong Q, Rimer JM, Bidani SS, Gale SE, Goree M, Hu E, Lanik WE, Huang E, Bando JK, Liu V, Lewis AN, Bustos A, Hodzic Z, Laury ML, Good M. Indole-3-Carbinol-Dependent Aryl Hydrocarbon Receptor Signaling Attenuates the Inflammatory Response in Experimental Necrotizing Enterocolitis. Immunohorizons 2021; 5:193-209. [PMID: 33906960 PMCID: PMC8173979 DOI: 10.4049/immunohorizons.2100018] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 03/01/2021] [Indexed: 01/10/2023] Open
Abstract
Necrotizing enterocolitis (NEC) causes significant morbidity and mortality in premature infants; therefore, the identification of therapeutic and preventative strategies against NEC remains a high priority. The ligand-dependent transcription factor aryl hydrocarbon receptor (AhR) is well known to contribute to the regulation of intestinal microbial communities and amelioration of intestinal inflammation. However, the role of AhR signaling in NEC is unclear. Experimental NEC was induced in 4-d-old wild-type mice or mice lacking AhR expression in the intestinal epithelial cells or AhR expression in CD11c+ cells (AhRΔCD11c) by subjecting animals to twice daily hypoxic stress and gavage feeding with formula supplemented with LPS and enteric bacteria. During NEC, compared with wild-type mice treated with vehicle, littermates treated with an AhR proligand, indole-3-carbinol, had reduced expression of Il1b and Marco, a scavenger receptor that mediates dendritic cell activation and the recognition and clearance of bacterial pathogens by macrophages. Furthermore, indole-3-carbinol treatment led to the downregulation of genes involved in cytokine and chemokine, as revealed by pathway enrichment analysis. AhR expression in the intestinal epithelial cells and their cre-negative mouse littermates were similarly susceptible to experimental NEC, whereas AhRΔCD11c mice with NEC exhibited heightened inflammatory responses compared with their cre-negative mouse littermates. In seeking to determine the mechanisms involved in this increased inflammatory response, we identified the Tim-4- monocyte-dependent subset of macrophages as increased in AhRΔCD11c mice compared with their cre-negative littermates. Taken together, these findings demonstrate the potential for AhR ligands as a novel immunotherapeutic approach to the management of this devastating disease.
Collapse
Affiliation(s)
- Lila S Nolan
- Division of Newborn Medicine, Washington University School of Medicine, St. Louis, MO
| | - Belgacem Mihi
- Division of Newborn Medicine, Washington University School of Medicine, St. Louis, MO
| | | | - Qingqing Gong
- Division of Newborn Medicine, Washington University School of Medicine, St. Louis, MO
| | - Jamie M Rimer
- Division of Newborn Medicine, Washington University School of Medicine, St. Louis, MO
| | - Shay S Bidani
- Division of Newborn Medicine, Washington University School of Medicine, St. Louis, MO
| | - Sarah E Gale
- Division of Newborn Medicine, Washington University School of Medicine, St. Louis, MO
| | - Martin Goree
- Division of Newborn Medicine, Washington University School of Medicine, St. Louis, MO
| | - Elise Hu
- Division of Newborn Medicine, Washington University School of Medicine, St. Louis, MO
| | - Wyatt E Lanik
- Division of Newborn Medicine, Washington University School of Medicine, St. Louis, MO
| | - Elizabeth Huang
- Division of Newborn Medicine, Washington University School of Medicine, St. Louis, MO
| | - Jennifer K Bando
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Victoria Liu
- Washington University in St. Louis, St. Louis, MO
| | - Angela N Lewis
- Division of Newborn Medicine, Washington University School of Medicine, St. Louis, MO
| | - Aiza Bustos
- Washington University in St. Louis, St. Louis, MO
| | - Zerina Hodzic
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA; and
| | - Marie L Laury
- Genome Technology Access Center, McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO
| | - Misty Good
- Division of Newborn Medicine, Washington University School of Medicine, St. Louis, MO;
| |
Collapse
|
21
|
Westfall S, Caracci F, Zhao D, Wu QL, Frolinger T, Simon J, Pasinetti GM. Microbiota metabolites modulate the T helper 17 to regulatory T cell (Th17/Treg) imbalance promoting resilience to stress-induced anxiety- and depressive-like behaviors. Brain Behav Immun 2021; 91:350-368. [PMID: 33096252 PMCID: PMC7986984 DOI: 10.1016/j.bbi.2020.10.013] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/07/2020] [Accepted: 10/10/2020] [Indexed: 02/08/2023] Open
Abstract
Chronic stress disrupts immune homeostasis while gut microbiota-derived metabolites attenuate inflammation, thus promoting resilience to stress-induced immune and behavioral abnormalities. There are both peripheral and brain region-specific maladaptations of the immune response to chronic stress that produce interrelated mechanistic considerations required for the design of novel therapeutic strategies for prevention of stress-induced psychological impairment. This study shows that a combination of probiotics and polyphenol-rich prebiotics, a synbiotic, attenuates the chronic-stress induced inflammatory responses in the ileum and the prefrontal cortex promoting resilience to the consequent depressive- and anxiety-like behaviors in male mice. Pharmacokinetic studies revealed that this effect may be attributed to specific synbiotic-produced metabolites including 4-hydroxyphenylpropionic, 4-hydroxyphenylacetic acid and caffeic acid. Using a model of chronic unpredictable stress, behavioral abnormalities were associated to strong immune cell activation and recruitment in the ileum while inflammasome pathways were implicated in the prefrontal cortex and hippocampus. Chronic stress also upregulated the ratio of activated proinflammatory T helper 17 (Th17) to regulatory T cells (Treg) in the liver and ileum and it was predicted with ingenuity pathway analysis that the aryl hydrocarbon receptor (AHR) could be driving the synbiotic's effect on the ileum's inflammatory response to stress. Synbiotic treatment indiscriminately attenuated the stress-induced immune and behavioral aberrations in both the ileum and the brain while in a gut-immune co-culture model, the synbiotic-specific metabolites promoted anti-inflammatory activity through the AHR. Overall, this study characterizes a novel synbiotic treatment for chronic-stress induced behavioral impairments while defining a putative mechanism of gut-microbiota host interaction for modulating the peripheral and brain immune systems.
Collapse
Affiliation(s)
- Susan Westfall
- Icahn School of Medicine at Mount Sinai, Department of Neurology, New York, NY, USA
| | - Francesca Caracci
- Icahn School of Medicine at Mount Sinai, Department of Neurology, New York, NY, USA
| | - Danyue Zhao
- Department of Plant Biology, Rutgers University, New Brunswick, NJ, USA
| | - Qing-li Wu
- Department of Plant Biology, Rutgers University, New Brunswick, NJ, USA
| | - Tal Frolinger
- Icahn School of Medicine at Mount Sinai, Department of Neurology, New York, NY, USA
| | - James Simon
- Department of Plant Biology, Rutgers University, New Brunswick, NJ, USA
| | - Giulio Maria Pasinetti
- Icahn School of Medicine at Mount Sinai, Department of Neurology, New York, NY, USA; Geriatric Research, Education and Clinical Center, James J. Peters Veterans Affairs Medical Center, Bronx, NY, USA.
| |
Collapse
|
22
|
Han H, Jayaraman A, Safe S, Chapkin RS. Targeting the aryl hydrocarbon receptor in stem cells to improve the use of food as medicine. CURRENT STEM CELL REPORTS 2020; 6:109-118. [PMID: 34395177 PMCID: PMC8362759 DOI: 10.1007/s40778-020-00184-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2020] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Intestinal stem cells, the most rapidly proliferating adult stem cells, are exquisitely sensitive to extrinsic dietary factors. Uncontrolled regulation of intestinal stem cells is closely linked to colon tumorigenesis. This review focuses on how dietary and microbial derived cues regulate intestinal stem cell functionality and colon tumorigenesis in mouse models by targeting the aryl hydrocarbon receptor (AhR). RECENT FINDINGS AhR, a ligand activated transcription factor, can integrate environmental, dietary and microbial cues to modulate intestinal stem cell proliferation, differentiation and their microenvironment, affecting colon cancer risk. Modulation of AhR activity is associated with many chronic diseases, including inflammatory bowel diseases where AhR expression is protective. SUMMARY AhR signaling controls the maintenance and differentiation of intestinal stem cells, influences local niche factors, and plays a protective role in colon tumorigenesis. Mounting evidence suggests that extrinsic nutritional/dietary cues which modulate AhR signaling may be a promising approach to colon cancer chemoprevention.
Collapse
Affiliation(s)
- Huajun Han
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX, 77843
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX, 77843
| | - Arul Jayaraman
- Department of Chemical Engineering, Texas A&M University, College Station, TX, 77843
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, 77843
| | - Robert S. Chapkin
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX, 77843
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX, 77843
- Department of Nutrition, Texas A&M University, College Station, TX, 77843
| |
Collapse
|
23
|
Dong F, Hao F, Murray IA, Smith PB, Koo I, Tindall AM, Kris-Etherton PM, Gowda K, Amin SG, Patterson AD, Perdew GH. Intestinal microbiota-derived tryptophan metabolites are predictive of Ah receptor activity. Gut Microbes 2020; 12:1-24. [PMID: 32783770 PMCID: PMC7524359 DOI: 10.1080/19490976.2020.1788899] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Commensal microbiota-dependent tryptophan catabolism within the gastrointestinal tract is known to exert profound effects upon host physiology, including the maintenance of epithelial barrier and immune function. A number of abundant microbiota-derived tryptophan metabolites exhibit activation potential for the aryl hydrocarbon receptor (AHR). Gene expression facilitated by AHR activation through the presence of dietary or microbiota-generated metabolites can influence gastrointestinal homeostasis and confer protection from intestinal challenges. Utilizing untargeted mass spectrometry-based metabolomics profiling, combined with AHR activity screening assays, we identify four previously unrecognized tryptophan metabolites, present in mouse cecal contents and human stool, with the capacity to activate AHR. Using GC/MS and LC/MS platforms, quantification of these novel AHR activators, along with previously established AHR-activating tryptophan metabolites, was achieved, providing a relative order of abundance. Using physiologically relevant concentrations and quantitative gene expression analyses, the relative efficacy of these tryptophan metabolites with regard to mouse or human AHR activation potential is examined. These data reveal indole, 2-oxindole, indole-3-acetic acid and kynurenic acid as the dominant AHR activators in mouse cecal contents and human stool from participants on a controlled diet. Here we provide the first documentation of the relative abundance and AHR activation potential of a panel of microbiota-derived tryptophan metabolites. Furthermore, these data reveal the human AHR to be more sensitive, at physiologically relevant concentrations, to tryptophan metabolite activation than mouse AHR. Additionally, correlation analyses indicate a relationship linking major tryptophan metabolite abundance with AHR activity, suggesting these cecal/fecal metabolites represent biomarkers of intestinal AHR activity.
Collapse
Affiliation(s)
- Fangcong Dong
- Department of Veterinary and Biomedical Sciences and the Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, USA
| | - Fuhua Hao
- Department of Veterinary and Biomedical Sciences and the Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, USA
| | - Iain A. Murray
- Department of Veterinary and Biomedical Sciences and the Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, USA
| | - Philip B. Smith
- The Huck Institutes of the Life Sciences, the Pennsylvania State University, University Park, PA, USA
| | - Imhoi Koo
- Department of Veterinary and Biomedical Sciences and the Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, USA
| | - Alyssa M. Tindall
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Penny M. Kris-Etherton
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Krishne Gowda
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - Shantu G. Amin
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - Andrew D. Patterson
- Department of Veterinary and Biomedical Sciences and the Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, USA
| | - Gary H. Perdew
- Department of Veterinary and Biomedical Sciences and the Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, USA,CONTACT Gary H. Perdew Department of Veterinary and Biomedical Sciences and the Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
24
|
Han H, Davidson LA, Fan Y, Goldsby JS, Yoon G, Jin U, Wright GA, Landrock KK, Weeks BR, Wright RC, Allred CD, Jayaraman A, Ivanov I, Roper J, Safe SH, Chapkin RS. Loss of aryl hydrocarbon receptor potentiates FoxM1 signaling to enhance self-renewal of colonic stem and progenitor cells. EMBO J 2020; 39:e104319. [PMID: 32915464 PMCID: PMC7527924 DOI: 10.15252/embj.2019104319] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 06/28/2020] [Accepted: 07/02/2020] [Indexed: 12/26/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor that senses xenobiotics, diet, and gut microbial-derived metabolites, is increasingly recognized as a key regulator of intestinal biology. However, its effects on the function of colonic stem and progenitor cells remain largely unexplored. Here, we observed that inducible deletion of AhR in Lgr5+ stem cells increases the percentage of colonic stem cells and enhances organoid initiating capacity and growth of sorted stem and progenitor cells, while AhR activation has the opposite effect. Moreover, intestinal-specific AhR knockout increases basal stem cell and crypt injury-induced cell proliferation and promotes colon tumorigenesis in a preclinical colitis-associated tumor model by upregulating FoxM1 signaling. Mechanistically, AhR transcriptionally suppresses FoxM1 expression. Activation of AhR in human organoids recapitulates phenotypes observed in mice, such as reduction in the percentage of colonic stem cells, promotion of stem cell differentiation, and attenuation of FoxM1 signaling. These findings indicate that the AhR-FoxM1 axis, at least in part, mediates colonic stem/progenitor cell behavior.
Collapse
Affiliation(s)
- Huajun Han
- Program in Integrative Nutrition and Complex DiseasesTexas A&M UniversityCollege StationTXUSA
- Department of Biochemistry & BiophysicsTexas A&M UniversityCollege StationTXUSA
| | - Laurie A Davidson
- Program in Integrative Nutrition and Complex DiseasesTexas A&M UniversityCollege StationTXUSA
- Department of NutritionTexas A&M UniversityCollege StationTXUSA
| | - Yang‐Yi Fan
- Program in Integrative Nutrition and Complex DiseasesTexas A&M UniversityCollege StationTXUSA
- Department of NutritionTexas A&M UniversityCollege StationTXUSA
| | - Jennifer S Goldsby
- Program in Integrative Nutrition and Complex DiseasesTexas A&M UniversityCollege StationTXUSA
- Department of NutritionTexas A&M UniversityCollege StationTXUSA
| | - Grace Yoon
- Department of StatisticsTexas A&M UniversityCollege StationTXUSA
| | - Un‐Ho Jin
- Veterinary Physiology and PharmacologyTexas A&M UniversityCollege StationTXUSA
| | - Gus A Wright
- Department of Veterinary PathobiologyTexas A&M UniversityCollege StationTXUSA
| | - Kerstin K Landrock
- Program in Integrative Nutrition and Complex DiseasesTexas A&M UniversityCollege StationTXUSA
- Department of NutritionTexas A&M UniversityCollege StationTXUSA
| | - Bradley R Weeks
- Department of Veterinary PathobiologyTexas A&M UniversityCollege StationTXUSA
| | - Rachel C Wright
- Program in Integrative Nutrition and Complex DiseasesTexas A&M UniversityCollege StationTXUSA
- Department of NutritionTexas A&M UniversityCollege StationTXUSA
| | | | - Arul Jayaraman
- Department of Chemical EngineeringTexas A&M UniversityCollege StationTXUSA
| | - Ivan Ivanov
- Program in Integrative Nutrition and Complex DiseasesTexas A&M UniversityCollege StationTXUSA
- Veterinary Physiology and PharmacologyTexas A&M UniversityCollege StationTXUSA
| | - Jatin Roper
- Department of MedicineDivision of GastroenterologyDuke University School of MedicineDurhamNCUSA
| | - Stephen H Safe
- Veterinary Physiology and PharmacologyTexas A&M UniversityCollege StationTXUSA
| | - Robert S Chapkin
- Program in Integrative Nutrition and Complex DiseasesTexas A&M UniversityCollege StationTXUSA
- Department of Biochemistry & BiophysicsTexas A&M UniversityCollege StationTXUSA
- Department of NutritionTexas A&M UniversityCollege StationTXUSA
| |
Collapse
|
25
|
Manzella CR, Jayawardena D, Pagani W, Li Y, Alrefai WA, Bauer J, Jung B, Weber CR, Gill RK. Serum Serotonin Differentiates Between Disease Activity States in Crohn's Patients. Inflamm Bowel Dis 2020; 26:1607-1618. [PMID: 32844174 PMCID: PMC7500525 DOI: 10.1093/ibd/izaa208] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Diagnosis and monitoring of inflammatory bowel diseases (IBDs) utilize invasive methods including endoscopy and tissue biopsy, with blood tests being less specific for IBDs. Substantial evidence has implicated involvement of the neurohormone serotonin (5-hydroxytryptamine, 5-HT) in the pathophysiology of IBDs. The current study investigated whether serum 5-HT is elevated in patients with active ulcerative colitis (UC) or Crohn's disease (CD). METHODS Serum samples were obtained from a German cohort of 96 CD and UC patients with active disease, refractory disease, or remission of disease based upon their disease activity index (DAI) and disease history. High pressure liquid chromatography with tandemmass spectrometry was used to measure 5-HT, tryptophan (TRP), and kynurenine (KYN) levels in the serum samples, and Luminex Multiplex ELISA was used to measure cytokine levels. Intestinal mucosal biopsies were obtained from a separate cohort of healthy and CD patients, and the immunoreactivity of the serotonin transporter (SERT) was determined. RESULTS There was no statistically significant difference in TRP or KYN levels between disease categories in either UC or CD. Interestingly, 5-HT levels were significantly elevated in patients with active CD but not active UC when compared with the levels in remission or refractory disease. Serum 5-HT was superior to C-reactive protein and circulating cytokines in differentiating between disease categories in CD. Additionally, SERT immunoreactivity was decreased in the ileum and colon of patients with CD compared to healthy controls. CONCLUSION We have shown that the serum 5-HT can differentiate between active disease and refractory disease or remission among CD patients, emphasizing the potential suitability of serum 5-HT as an auxiliary measure in diagnosing active CD.
Collapse
Affiliation(s)
- Christopher R Manzella
- Department of Physiology & Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Dulari Jayawardena
- Division of Gastroenterology & Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Wilfredo Pagani
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Ye Li
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Waddah A Alrefai
- Department of Physiology & Biophysics, University of Illinois at Chicago, Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Jessica Bauer
- Department of Medicine, University of Washington, WA, USA
| | - Barbara Jung
- Department of Medicine, University of Washington, WA, USA
| | | | - Ravinder K Gill
- Department of Physiology & Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
26
|
Uebanso T, Shimohata T, Mawatari K, Takahashi A. Functional Roles of B‐Vitamins in the Gut and Gut Microbiome. Mol Nutr Food Res 2020; 64:e2000426. [DOI: 10.1002/mnfr.202000426] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/31/2020] [Indexed: 01/19/2023]
Affiliation(s)
- Takashi Uebanso
- Department of Preventive Environment and Nutrition, Institute of Biomedical Sciences Tokushima University Graduate School Tokushima 770–8503 Japan
| | - Takaaki Shimohata
- Department of Preventive Environment and Nutrition, Institute of Biomedical Sciences Tokushima University Graduate School Tokushima 770–8503 Japan
| | - Kazuaki Mawatari
- Department of Preventive Environment and Nutrition, Institute of Biomedical Sciences Tokushima University Graduate School Tokushima 770–8503 Japan
| | - Akira Takahashi
- Department of Preventive Environment and Nutrition, Institute of Biomedical Sciences Tokushima University Graduate School Tokushima 770–8503 Japan
| |
Collapse
|
27
|
Microbial tryptophan metabolites regulate gut barrier function via the aryl hydrocarbon receptor. Proc Natl Acad Sci U S A 2020; 117:19376-19387. [PMID: 32719140 DOI: 10.1073/pnas.2000047117] [Citation(s) in RCA: 374] [Impact Index Per Article: 74.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Inflammatory bowel diseases (IBDs), including Crohn's disease and ulcerative colitis, are associated with dysbiosis of the gut microbiome. Emerging evidence suggests that small-molecule metabolites derived from bacterial breakdown of a variety of dietary nutrients confer a wide array of host benefits, including amelioration of inflammation in IBDs. Yet, in many cases, the molecular pathways targeted by these molecules remain unknown. Here, we describe roles for three metabolites-indole-3-ethanol, indole-3-pyruvate, and indole-3-aldehyde-which are derived from gut bacterial metabolism of the essential amino acid tryptophan, in regulating intestinal barrier function. We determined that these metabolites protect against increased gut permeability associated with a mouse model of colitis by maintaining the integrity of the apical junctional complex and its associated actin regulatory proteins, including myosin IIA and ezrin, and that these effects are dependent on the aryl hydrocarbon receptor. Our studies provide a deeper understanding of how gut microbial metabolites affect host defense mechanisms and identify candidate pathways for prophylactic and therapeutic treatments for IBDs.
Collapse
|
28
|
Does NLRP3 Inflammasome and Aryl Hydrocarbon Receptor Play an Interlinked Role in Bowel Inflammation and Colitis-Associated Colorectal Cancer? Molecules 2020; 25:molecules25102427. [PMID: 32456012 PMCID: PMC7287590 DOI: 10.3390/molecules25102427] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/17/2020] [Accepted: 05/21/2020] [Indexed: 12/22/2022] Open
Abstract
Inflammation is a hallmark in many forms of cancer; with colitis-associated colorectal cancer (CAC) being a progressive intestinal inflammation due to inflammatory bowel disease (IBD). While this is an exemplification of the negatives of inflammation, it is just as crucial to have some degree of the inflammatory process to maintain a healthy immune system. A pivotal component in the maintenance of such intestinal homeostasis is the innate immunity component, inflammasomes. Inflammasomes are large, cytosolic protein complexes formed following stimulation of microbial and stress signals that lead to the expression of pro-inflammatory cytokines. The NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome has been extensively studied in part due to its strong association with colitis and CAC. The aryl hydrocarbon receptor (AhR) has recently been acknowledged for its connection to the immune system aside from its role as an environmental sensor. AhR has been described to play a role in the inhibition of the NLRP3 inflammasome activation pathway. This review will summarise the signalling pathways of both the NLRP3 inflammasome and AhR; as well as new-found links between these two signalling pathways in intestinal immunity and some potential therapeutic agents that have been found to take advantage of this link in the treatment of colitis and CAC.
Collapse
|
29
|
The aryl hydrocarbon receptor: an environmental sensor integrating immune responses in health and disease. Nat Rev Immunol 2019; 19:184-197. [PMID: 30718831 DOI: 10.1038/s41577-019-0125-8] [Citation(s) in RCA: 778] [Impact Index Per Article: 129.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The environment, diet, microbiota and body's metabolism shape complex biological processes in health and disease. However, our understanding of the molecular pathways involved in these processes is still limited. The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that integrates environmental, dietary, microbial and metabolic cues to control complex transcriptional programmes in a ligand-specific, cell-type-specific and context-specific manner. In this Review, we summarize our current knowledge of AHR and the transcriptional programmes it controls in the immune system. Finally, we discuss the role of AHR in autoimmune and neoplastic diseases of the central nervous system, with a special focus on the gut immune system, the gut-brain axis and the therapeutic potential of targeting AHR in neurological disorders.
Collapse
|
30
|
Miao Y, Lv Q, Qiao S, Yang L, Tao Y, Yan W, Wang P, Cao N, Dai Y, Wei Z. Alpinetin improves intestinal barrier homeostasis via regulating AhR/suv39h1/TSC2/mTORC1/autophagy pathway. Toxicol Appl Pharmacol 2019; 384:114772. [PMID: 31676321 DOI: 10.1016/j.taap.2019.114772] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 09/27/2019] [Accepted: 10/01/2019] [Indexed: 02/08/2023]
Abstract
The injury of intestinal epithelial barrier is considered as the key pathophysiological process in response to gastrointestinal infection and inflammation, and plays an important role in the initiation and development of colitis. Alpinetin has been shown to improve intestinal barrier homeostasis under colitis condition, but the mechanism is still unclear. Here, we showed that alpinetin significantly improved transepithelial electrical resistance (TEER) in TNF-α-stimulated Caco-2 cells, which was mainly mediated by inhibiting the apoptosis. Mechanistic studies demonstrated that alpinetin markedly increased the production of autophagosomes, along with obvious regulation of LC3B-II, beclin-1, p62, Atg7 and Atg5 expressions. In addition, it also markedly repressed the activation of mTORC1 signaling pathway, which was ascribed to TSC2 rather than p-AKT, p-ERK, p-AMPKα or PTEN expressions in Caco-2 and NCM460 cells. Furthermore, the enrichment of H3K9me3 at TSC2 promoter region was decreased and ubiquitin proteasome degradation of suv39h1 was increased. Additionally, alpinetin activated aryl hydrocarbon receptor (AhR) and promoted co-localization of AhR with suv39h1 in the cytoplasm. The relationship between alpinetin-regulated AhR/suv39h1/TSC2/mTORC1 signals, autophagy and apoptosis of Caco-2 and NCM460 cells was confirmed by using CH223191, siAhR, siTSC2 and chloroquine. Finally, CH223191 and leucine abolished alpinetin-mediated inhibition of intestinal epithelial cells apoptosis, improvement of intestinal epithelial barrier and amelioration of colitis.
Collapse
Affiliation(s)
- Yumeng Miao
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Qi Lv
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Simiao Qiao
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Ling Yang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Yu Tao
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Wenxin Yan
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Pengfei Wang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Na Cao
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Yue Dai
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China.
| | - Zhifeng Wei
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China.
| |
Collapse
|
31
|
Muku GE, Murray IA, Perdew GH. Activation of the Ah Receptor Modulates Gastrointestinal Homeostasis and the Intestinal Microbiome. ACTA ACUST UNITED AC 2019. [DOI: 10.1007/s40495-019-00197-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
32
|
Ji J, Qu H. Cross-regulatory Circuit Between AHR and Microbiota. Curr Drug Metab 2019; 20:4-8. [PMID: 29380692 DOI: 10.2174/1389200219666180129151150] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 10/12/2017] [Accepted: 11/26/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND The gut microbes have a close symbiotic relationship with their host. Interactions between host and the microbiota affect the nutritional, immunological, and physiological status of the host. The Aryl Hydrocarbon Receptor (AHR) is a ligand activated transcription factor that mediates the toxicity of xenobiotics. Recently, the relationship between the gut microbiota and AHR has attracted the attention of many researchers. METHODS We undertook a structured search of bibliographic databases for peer-reviewed research literature. RESULTS We found and reviewed 49 peer-reviewed papers dealing with the major aspects related to the crosstalk between AHR and microbiota. The AHR influences the intestinal microbiota population and mediates host-microbe homeostasis. Interestingly, the gut microbiota also produces ligands of AHR from bacterial metabolism and thereby activates the AHR signaling pathway. Concusion: This review presents current knowledge of the cross-regulatory circuit between the AHR and intestinal microbiota. The findings of this review confirm the importance of AHR-microbiota interactions in health and disease.
Collapse
Affiliation(s)
- Jian Ji
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Hao Qu
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| |
Collapse
|
33
|
Li Y, Xie HQ, Zhang W, Wei Y, Sha R, Xu L, Zhang J, Jiang Y, Guo TL, Zhao B. Type 3 innate lymphoid cells are altered in colons of C57BL/6 mice with dioxin exposure. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 662:639-645. [PMID: 30703721 DOI: 10.1016/j.scitotenv.2019.01.139] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 01/10/2019] [Accepted: 01/12/2019] [Indexed: 06/09/2023]
Abstract
Type 3 innate lymphoid cells (ILC3s) are distributed in the gut and regulate inflammation by secreting cytokines, including interferon (IFN)-γ and interleukin (IL)-17. The maintenance and function of ILC3s involve the activity of aryl hydrocarbon receptor (AhR), a potent ligand of which is 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), one of the most toxic dioxin congeners. Thus, TCDD exposure might affect ILC3s. To obtain in vivo evidence supporting this notion, we exposed female C57BL/6 mice orally to TCDD (low/high doses: 0.1/10 μg/kg body weight) during pregnancy and lactation periods, and after the exposure, evaluated the mothers and offspring for alterations in ILC3 differentiation and function in the colon. ILC3 frequency among colonic lamina propria lymphocytes was preferentially diminished in the offspring, and, in parallel, the median fluorescence intensity (MFI) of retinoic acid receptor-related orphan receptor (ROR)γt, which is associated with ILC3 differentiation, was also decreased in ILC3s. Conversely, the percentages of two subsets of the cells, one positive for natural cytotoxicity receptor NKp46 and the other for IL-17a, were increased in TCDD-exposed mothers and offspring. Moreover, the percentage of IFN-γ+ ILC3s was increased specifically in the mothers, but this was in conjunction with a significant decrease in the MFI of IFN-γ, which suggests that the IFN-γ+ ILC3 subset was functionally altered. In conclusion, maternal exposure to TCDD suppresses ILC3 differentiation in the offspring and influences ILC3 function in distinct manners in the mother and offspring. Our study provides new insights into the intergenerational interference of dioxins in colonic ILC3s.
Collapse
Affiliation(s)
- Yunping Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center of Eco-Environment Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Heidi Qunhui Xie
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center of Eco-Environment Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wanglong Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center of Eco-Environment Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yunbo Wei
- Laboratory of Immunology for Environment and Health, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Rui Sha
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center of Eco-Environment Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Li Xu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center of Eco-Environment Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jianqing Zhang
- Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Yousheng Jiang
- Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Tai L Guo
- Department of Veterinary Biosciences and Diagnostic Imaging, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Bin Zhao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center of Eco-Environment Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
34
|
Uehara D, Tojima H, Kakizaki S, Yamazaki Y, Horiguchi N, Takizawa D, Sato K, Yamada M, Uraoka T. Constitutive androstane receptor and pregnane X receptor cooperatively ameliorate DSS-induced colitis. Dig Liver Dis 2019; 51:226-235. [PMID: 30442521 DOI: 10.1016/j.dld.2018.10.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 10/06/2018] [Accepted: 10/09/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Nuclear receptor pregnane X receptor (PXR) was shown to be protective in case of dextran sulfate sodium (DSS)-induced colitis. Constitutive androstane receptor (CAR) belongs to the same nuclear receptor subfamily with PXR. The roles of both receptors in DSS-induced colitis were evaluated. METHODS Wild-type, Car-null, Pxr-null, and Car/Pxr-null mice were treated with a CAR/PXR agonist or vehicle and administered 2.5% DSS in the drinking water. The typical clinical symptoms, histological scoring, proinflammatory cytokine, and apoptosis were analyzed. RESULTS Mice treated with the PXR agonist pregnenolone-16α-carbonitrile (PCN) were protected from DSS-induced colitis, as in a previous study. Mice treated with the CAR agonist, 4-bis[2-(3,5-dichloropyridyloxy)]benzene (TCPOBOP) were also protected from DSS-induced colitis. Interestingly, the protective effects of PCN in the Car-null mice and those of TCPOBOP in the Pxr-null mice both decreased. PCN or TCPOBOP pretreatment significantly decreased the macrophage and monocyte infiltration in DSS-induced colitis. PXR and CAR agonists reduced the mRNA expression of several proinflammatory cytokines in a PXR- and CAR-dependent manner, respectively. CAR inhibited apoptosis by inducing Gadd45b. PXR inhibited TNF-α and IL-1b and CAR induced Gadd45b in in vitro cell analyses. CONCLUSIONS We showed that CAR and PXR cooperatively ameliorate DSS-induced colitis. PXR and CAR protected against DSS-induced colitis by inhibiting proinflammatory cytokines and apoptosis, respectively.
Collapse
Affiliation(s)
- Daisuke Uehara
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan; Department of Gastroenterology and Hepatology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Hiroki Tojima
- Department of Gastroenterology and Hepatology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Satoru Kakizaki
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan; Department of Gastroenterology and Hepatology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan.
| | - Yuichi Yamazaki
- Department of Gastroenterology and Hepatology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Norio Horiguchi
- Department of Gastroenterology and Hepatology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Daichi Takizawa
- Department of Gastroenterology and Hepatology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Ken Sato
- Department of Gastroenterology and Hepatology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Masanobu Yamada
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Toshio Uraoka
- Department of Gastroenterology and Hepatology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| |
Collapse
|
35
|
Lamas B, Natividad JM, Sokol H. Aryl hydrocarbon receptor and intestinal immunity. Mucosal Immunol 2018; 11:1024-1038. [PMID: 29626198 DOI: 10.1038/s41385-018-0019-2] [Citation(s) in RCA: 363] [Impact Index Per Article: 51.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Revised: 02/24/2018] [Accepted: 02/26/2018] [Indexed: 02/04/2023]
Abstract
Aryl hydrocarbon receptor (AhR) is a member of the basic helix-loop-helix-(bHLH) superfamily of transcription factors, which are associated with cellular responses to environmental stimuli, such as xenobiotics and oxygen levels. Unlike other members of bHLH, AhR is the only bHLH transcription factor that is known to be ligand activated. Early AhR studies focused on understanding the role of AhR in mediating the toxicity and carcinogenesis properties of the prototypic ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). In recent years, however, it has become apparent that, in addition to its toxicological involvement, AhR is highly receptive to a wide array of endogenous and exogenous ligands, and that its activation leads to a myriad of key host physiological functions. In this study, we review the current understanding of the functions of AhR in the mucosal immune system with a focus on its role in intestinal barrier function and intestinal immune cells, as well as in intestinal homeostasis.
Collapse
Affiliation(s)
- Bruno Lamas
- Laboratoire de biomolécules, LBM, Sorbonne Université, École normale supérieure, PSL Research University, CNRS, INSERM, AP-HP, Hôpital Saint-Antoine, Paris, F-75005, France.,Micalis Institute, Institut National de la Recherche Agronomique (INRA), AgroParisTech, Université Paris-Saclay, Jouy en Josas, 78350, France
| | - Jane M Natividad
- Micalis Institute, Institut National de la Recherche Agronomique (INRA), AgroParisTech, Université Paris-Saclay, Jouy en Josas, 78350, France
| | - Harry Sokol
- Laboratoire de biomolécules, LBM, Sorbonne Université, École normale supérieure, PSL Research University, CNRS, INSERM, AP-HP, Hôpital Saint-Antoine, Paris, F-75005, France. .,Micalis Institute, Institut National de la Recherche Agronomique (INRA), AgroParisTech, Université Paris-Saclay, Jouy en Josas, 78350, France.
| |
Collapse
|
36
|
Cannon AR, Kuprys PV, Cobb AN, Ding X, Kothari AN, Kuo PC, Eberhardt JM, Hammer AM, Morris NL, Li X, Choudhry MA. Alcohol enhances symptoms and propensity for infection in inflammatory bowel disease patients and a murine model of DSS-induced colitis. J Leukoc Biol 2018; 104:543-555. [PMID: 29775230 DOI: 10.1002/jlb.4ma1217-506r] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/28/2018] [Accepted: 04/11/2018] [Indexed: 12/13/2022] Open
Abstract
Over 1.4 million Americans have been diagnosed with inflammatory bowel disease (IBD), and ulcerative colitis (UC) makes up approximately half of those diagnoses. As a disease, UC cycles between periods of remission and flare, which is characterized by intense abdominal pain, increased weight loss, intestinal inflammation, rectal bleeding, and dehydration. Interestingly, a widespread recommendation to IBD patients for avoidance of a flare period is "Don't Drink Alcohol" as recent work correlated alcohol consumption with increased GI symptoms in patients with IBD. Alcohol alone not only induces a systemic pro-inflammatory response, but can also be directly harmful to gut barrier integrity. However, how alcohol could result in the exacerbation of UC in both patients and murine models of colitis has yet to be elucidated. Therefore, we conducted a retrospective analysis of patients admitted for IBD with a documented history of alcohol use in conjunction with a newly developed mouse model of binge alcohol consumption following dextran sulfate sodium (DSS)-induced colitis. We found that alcohol negatively impacts clinical outcomes of patients with IBD, specifically increased intestinal infections, antibiotic injections, abdomen CT scans, and large intestine biopsies. Furthermore, in our mouse model of binge alcohol consumption following an induced colitis flare, we found alcohol exacerbates weight loss, clinical scores, colonic shortening and inflammation, and propensity to infection. These findings highlight alcohol's ability to potentiate symptoms and susceptibility to infection in UC and suggest alcohol as an underlying factor in perpetuating symptoms of IBD.
Collapse
Affiliation(s)
- Abigail R Cannon
- Alcohol Research Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Burn & Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Integrative Cell Biology Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| | - Paulius V Kuprys
- Alcohol Research Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Burn & Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Integrative Cell Biology Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| | - Adrienne N Cobb
- Burn & Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Department of Surgery, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| | - Xianzhong Ding
- Department of Pathology, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| | - Anai N Kothari
- Burn & Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Department of Surgery, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| | - Paul C Kuo
- Department of Surgery, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| | - Joshua M Eberhardt
- Department of Surgery, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| | - Adam M Hammer
- Alcohol Research Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Burn & Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Integrative Cell Biology Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| | - Niya L Morris
- Alcohol Research Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Burn & Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Integrative Cell Biology Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| | - Xiaoling Li
- Alcohol Research Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Burn & Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Department of Surgery, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| | - Mashkoor A Choudhry
- Alcohol Research Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Burn & Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Department of Surgery, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Department of Pathology, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Department of Microbiology and Immunology, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Integrative Cell Biology Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| |
Collapse
|
37
|
Gao J, Xu K, Liu H, Liu G, Bai M, Peng C, Li T, Yin Y. Impact of the Gut Microbiota on Intestinal Immunity Mediated by Tryptophan Metabolism. Front Cell Infect Microbiol 2018; 8:13. [PMID: 29468141 PMCID: PMC5808205 DOI: 10.3389/fcimb.2018.00013] [Citation(s) in RCA: 829] [Impact Index Per Article: 118.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 01/12/2018] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota influences the health of the host, especially with regard to gut immune homeostasis and the intestinal immune response. In addition to serving as a nutrient enhancer, L-tryptophan (Trp) plays crucial roles in the balance between intestinal immune tolerance and gut microbiota maintenance. Recent discoveries have underscored that changes in the microbiota modulate the host immune system by modulating Trp metabolism. Moreover, Trp, endogenous Trp metabolites (kynurenines, serotonin, and melatonin), and bacterial Trp metabolites (indole, indolic acid, skatole, and tryptamine) have profound effects on gut microbial composition, microbial metabolism, the host's immune system, the host-microbiome interface, and host immune system-intestinal microbiota interactions. The aryl hydrocarbon receptor (AhR) mediates the regulation of intestinal immunity by Trp metabolites (as ligands of AhR), which is beneficial for immune homeostasis. Among Trp metabolites, AhR ligands consist of endogenous metabolites, including kynurenine, kynurenic acid, xanthurenic acid, and cinnabarinic acid, and bacterial metabolites, including indole, indole propionic acid, indole acetic acid, skatole, and tryptamine. Additional factors, such as aging, stress, probiotics, and diseases (spondyloarthritis, irritable bowel syndrome, inflammatory bowel disease, colorectal cancer), which are associated with variability in Trp metabolism, can influence Trp-microbiome-immune system interactions in the gut and also play roles in regulating gut immunity. This review clarifies how the gut microbiota regulates Trp metabolism and identifies the underlying molecular mechanisms of these interactions. Increased mechanistic insight into how the microbiota modulates the intestinal immune system through Trp metabolism may allow for the identification of innovative microbiota-based diagnostics, as well as appropriate nutritional supplementation of Trp to prevent or alleviate intestinal inflammation. Moreover, this review provides new insight regarding the influence of the gut microbiota on Trp metabolism. Additional comprehensive analyses of targeted Trp metabolites (including endogenous and bacterial metabolites) are essential for experimental preciseness, as the influence of the gut microbiota cannot be neglected, and may explain contradictory results in the literature.
Collapse
Affiliation(s)
- Jing Gao
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, China
- Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Kang Xu
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, China
- Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, China
| | - Hongnan Liu
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, China
- Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, China
| | - Gang Liu
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, China
- Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, China
| | - Miaomiao Bai
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, China
- Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, China
| | - Can Peng
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, China
- Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, China
| | - Tiejun Li
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, China
- Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, China
| | - Yulong Yin
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, China
- Key Laboratory of Agro-Ecology, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, China
- University of Chinese Academy of Sciences, Beijing, China
- College of Life Science, Hunan Normal University, Changsha, Hunan, China
| |
Collapse
|
38
|
Intestinal alkaline phosphatase ameliorates experimental colitis via toll-like receptor 4-dependent pathway. Eur J Pharmacol 2017; 820:156-166. [PMID: 29247612 DOI: 10.1016/j.ejphar.2017.12.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/07/2017] [Accepted: 12/08/2017] [Indexed: 12/31/2022]
Abstract
Intestinal alkaline phosphatase (IAP) is an intestinal brush border enzyme which plays an important role in gut homeostasis and mucosal inflammation. However, the mechanism of the protective effect of IAP is not fully elucidated. The aim of the present study was to evaluate whether the protective effect of IAP on colitis is mediated via the toll-like receptor 4 (TLR4)/nuclear factor-kappa B (NF-κB) pathway. For in vitro analysis, peritoneal macrophages from the wild-type (WT) and TLR4-deficient (TLR4-/-) C57BL/6 mice were used. IAP strongly inhibited the lipopolysaccharide (LPS)-induced production of tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) by suppressing the LPS-induced phosphorylation of IκBα and the DNA-binding activity of NF-κB in WT macrophages; however, the inhibitory effects mediated by IAP were reduced in the TLR4-/- macrophages. For in vivo analysis, the protective mechanisms of IAP on dextran sulfate sodium (DSS)-induced colitis were evaluated using WT and TLR4-/- mice. Although oral administration of IAP significantly attenuated the severity of colitis in both preventive and therapeutic models of WT mice, these protective effects were not significant in TLR4-/- mice. When immunohistochemical analysis of IκBα was performed in the colitic tissues, the inhibitory effects of IAP on IκBα phosphorylation were also observed in the colon of WT mice, but these effects decreased in the colon of TLR4-/- mice. In conclusion, the protective effects of IAP on colitis were mediated via the TLR4/NF-κB pathway. These results of this study shall be helpful in tailoring treatment against colitis using IAP.
Collapse
|
39
|
Spiljar M, Merkler D, Trajkovski M. The Immune System Bridges the Gut Microbiota with Systemic Energy Homeostasis: Focus on TLRs, Mucosal Barrier, and SCFAs. Front Immunol 2017; 8:1353. [PMID: 29163467 PMCID: PMC5670327 DOI: 10.3389/fimmu.2017.01353] [Citation(s) in RCA: 134] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 10/03/2017] [Indexed: 12/17/2022] Open
Abstract
The gut microbiota is essential for the development and regulation of the immune system and the metabolism of the host. Germ-free animals have altered immunity with increased susceptibility to immunologic diseases and show metabolic alterations. Here, we focus on two of the major immune-mediated microbiota-influenced components that signal far beyond their local environment. First, the activation or suppression of the toll-like receptors (TLRs) by microbial signals can dictate the tone of the immune response, and they are implicated in regulation of the energy homeostasis. Second, we discuss the intestinal mucosal surface is an immunologic component that protects the host from pathogenic invasion, is tightly regulated with regard to its permeability and can influence the systemic energy balance. The short chain fatty acids are a group of molecules that can both modulate the intestinal barrier and escape the gut to influence systemic health. As modulators of the immune response, the microbiota-derived signals influence functions of distant organs and can change susceptibility to metabolic diseases.
Collapse
Affiliation(s)
- Martina Spiljar
- Faculty of Medicine, Department of Cell Physiology and Metabolism, Centre Médical Universitaire, University of Geneva, Geneva, Switzerland.,Diabetes Center, Faculty of Medicine, Centre Médical Universitaire, University of Geneva, Geneva, Switzerland
| | - Doron Merkler
- Faculty of Medicine, Department of Pathology and Immunology, Centre Médical Universitaire, University of Geneva, Geneva, Switzerland
| | - Mirko Trajkovski
- Faculty of Medicine, Department of Cell Physiology and Metabolism, Centre Médical Universitaire, University of Geneva, Geneva, Switzerland.,Diabetes Center, Faculty of Medicine, Centre Médical Universitaire, University of Geneva, Geneva, Switzerland.,Institute of Genetics and Genomics in Geneva (iGE3), University of Geneva, Geneva, Switzerland
| |
Collapse
|
40
|
Hubbard TD, Murray IA, Nichols RG, Cassel K, Podolsky M, Kuzu G, Tian Y, Smith P, Kennett MJ, Patterson AD, Perdew GH. Dietary Broccoli Impacts Microbial Community Structure and Attenuates Chemically Induced Colitis in Mice in an Ah receptor dependent manner. J Funct Foods 2017; 37:685-698. [PMID: 29242716 PMCID: PMC5726276 DOI: 10.1016/j.jff.2017.08.038] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Consumption of broccoli mediates numerous chemo-protective benefits through the intake of phytochemicals, some of which modulate aryl hydrocarbon receptor (AHR) activity. Whether AHR activation is a critical aspect of the therapeutic potential of dietary broccoli is not known. Here we administered isocaloric diets, with or without supplementation of whole broccoli (15% w/w), to congenic mice expressing the high-affinity Ahrb/b or low-affinity Ahrd/d alleles, for 24 days and examined the effects on AHR activity, intestinal microbial community structure, inflammatory status, and response to chemically induced colitis. Cecal microbial community structure and metabolic potential were segregated according to host dietary and AHR status. Dietary broccoli associated with heightened intestinal AHR activity, decreased microbial abundance of the family Erysipelotrichaceae, and attenuation of colitis. In summary, broccoli consumption elicited an enhanced response in ligand-sensitive Ahrb/b mice, demonstrating that in part the beneficial aspects of dietary broccoli upon intestinal health are associated with heightened AHR activity.
Collapse
Affiliation(s)
- Troy D. Hubbard
- Department of Veterinary and Biomedical Sciences and The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802
| | - Iain A. Murray
- Department of Veterinary and Biomedical Sciences and The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802
| | - Robert G. Nichols
- Department of Veterinary and Biomedical Sciences and The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802
| | - Kaitlyn Cassel
- Department of Veterinary and Biomedical Sciences and The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802
| | - Michael Podolsky
- Department of Veterinary and Biomedical Sciences and The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802
| | - Guray Kuzu
- Center for Eukaryotic Gene Regulation, Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802
| | - Yuan Tian
- Department of Veterinary and Biomedical Sciences and The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802
| | - Phillip Smith
- The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802
| | - Mary J. Kennett
- Department of Veterinary and Biomedical Sciences and The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802
| | - Andrew D. Patterson
- Department of Veterinary and Biomedical Sciences and The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802
| | - Gary H. Perdew
- Department of Veterinary and Biomedical Sciences and The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802
| |
Collapse
|
41
|
Guo BJ, Bian ZX, Qiu HC, Wang YT, Wang Y. Biological and clinical implications of herbal medicine and natural products for the treatment of inflammatory bowel disease. Ann N Y Acad Sci 2017; 1401:37-48. [DOI: 10.1111/nyas.13414] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 05/15/2017] [Accepted: 05/18/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Bao-Jian Guo
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences; University of Macau; Taipa Macao SAR China
| | - Zhao-Xiang Bian
- School of Chinese Medicine and Hong Kong Chinese Medicine Study Centre; Hong Kong Baptist University; Kowloon Tong Hong Kong China
| | - Hong-Cong Qiu
- Guangxi Institute of Traditional Medical and Pharmaceutical Sciences and Guangxi Key Laboratory of Traditional Chinese Medicine Quality Standards; Nanning China
| | - Yi-Tao Wang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences; University of Macau; Taipa Macao SAR China
| | - Ying Wang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences; University of Macau; Taipa Macao SAR China
| |
Collapse
|
42
|
Kawai S, Iijima H, Shinzaki S, Hiyama S, Yamaguchi T, Araki M, Iwatani S, Shiraishi E, Mukai A, Inoue T, Hayashi Y, Tsujii M, Motooka D, Nakamura S, Iida T, Takehara T. Indigo Naturalis ameliorates murine dextran sodium sulfate-induced colitis via aryl hydrocarbon receptor activation. J Gastroenterol 2017; 52:904-919. [PMID: 27900483 DOI: 10.1007/s00535-016-1292-z] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 11/16/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND Indigo Naturalis (IN) is used as a traditional herbal medicine for ulcerative colitis (UC). However, the mechanisms of action of IN have not been clarified. We aimed to evaluate the efficacy of IN for ameliorating colonic inflammation. We further investigated the mechanisms of action of IN. METHODS Colitis severity was assessed in dextran sodium sulfate-induced colitis and trinitrobenzene sulfonic acid-induced colitis models with or without the oral administration of IN or indigo, which is a known major component of IN. Colonic lamina propria (LP) mononuclear cells isolated from IN-treated mice were analyzed with quantitative reverse transcription polymerase chain reaction (qRT-PCR) and flow cytometry. LP and splenic mononuclear cells cultured in vitro with IN or indigo were also analyzed. The role of the candidate receptor for indigo, the aryl hydrocarbon receptor (AhR), was analyzed using Ahr-deficient mice. RESULTS Colitis severity was significantly ameliorated in the IN and indigo treatment groups compared with the control group. The mRNA expression levels of interleukin (Il)-10 and Il-22 in the LP lymphocytes were increased by IN treatment. The treatment of splenocytes with IN or indigo increased the expression of anti-inflammatory cytokines and resulted in the expansion of IL-10-producing CD4+ T cells and IL-22-producing CD3-RORγt+ cells, but not CD4+Foxp3+ regulatory T cells. The amelioration of colitis by IN or indigo was abrogated in Ahr-deficient mice, in association with diminished regulatory cytokine production. CONCLUSIONS IN and indigo ameliorated murine colitis through AhR signaling activation, suggesting that AhR could be a promising therapeutic target for UC.
Collapse
MESH Headings
- Animals
- CD3 Complex/metabolism
- CD4 Lymphocyte Count
- CD4-Positive T-Lymphocytes/metabolism
- Cells, Cultured
- Colitis/chemically induced
- Colitis/drug therapy
- Colitis/pathology
- Dextran Sulfate
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/therapeutic use
- Female
- Forkhead Transcription Factors/metabolism
- Gene Expression/drug effects
- Indigo Carmine/pharmacology
- Indigo Carmine/therapeutic use
- Interleukin-10/genetics
- Interleukin-10/metabolism
- Interleukin-2 Receptor alpha Subunit/metabolism
- Interleukins/genetics
- Interleukins/metabolism
- Intestinal Mucosa/cytology
- Leukocytes, Mononuclear/metabolism
- Mice, Knockout
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- RNA, Messenger/metabolism
- Receptors, Aryl Hydrocarbon/deficiency
- Receptors, Aryl Hydrocarbon/drug effects
- Receptors, Aryl Hydrocarbon/genetics
- Receptors, Aryl Hydrocarbon/metabolism
- Severity of Illness Index
- Spleen/cytology
- T-Lymphocytes/metabolism
- T-Lymphocytes, Regulatory/metabolism
- Trinitrobenzenesulfonic Acid
- Interleukin-22
Collapse
Affiliation(s)
- Shoichiro Kawai
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hideki Iijima
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Shinichiro Shinzaki
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Satoshi Hiyama
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Inflammatory Bowel Disease, Osaka University Graduate School of Medicine, Suita, Japan
| | - Toshio Yamaguchi
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Manabu Araki
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Shuko Iwatani
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Eri Shiraishi
- Department of Gastroenterology and Hepatology, Sumitomo Hospital, Osaka, Japan
| | - Akira Mukai
- Department of Gastroenterology and Hepatology, Sumitomo Hospital, Osaka, Japan
| | - Takahiro Inoue
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshito Hayashi
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Masahiko Tsujii
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Gastroenterology and Hepatology, Higashiosaka City General Hospital, Higashiosaka, Japan
| | - Daisuke Motooka
- Department of Infection Metagenomics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Shota Nakamura
- Department of Infection Metagenomics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Tetsuya Iida
- Department of Infection Metagenomics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Tetsuo Takehara
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
43
|
Hudson GM, Flannigan KL, Erickson SL, Vicentini FA, Zamponi A, Hirota CL, Alston L, Altier C, Ghosh S, Rioux KP, Mani S, Chang TK, Hirota SA. Constitutive androstane receptor regulates the intestinal mucosal response to injury. Br J Pharmacol 2017; 174:1857-1871. [PMID: 28320072 DOI: 10.1111/bph.13787] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 02/21/2017] [Accepted: 03/14/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE The pathogenesis of the inflammatory bowel diseases (IBD), comprising Crohn's disease (CD) and ulcerative colitis (UC), involves aberrant interactions between a genetically susceptible individual, their microbiota and environmental factors. Alterations in xenobiotic receptor expression and function are associated with increased risk for IBD. Here, we have assessed the role of the constitutive androstane receptor (CAR), a xenobiotic receptor closely related to the pregnane X receptor, in the regulation of intestinal mucosal homeostasis. EXPERIMENTAL APPROACH CAR expression was assessed in intestinal mucosal biopsies obtained from CD and UC patients, and in C57/Bl6 mice exposed to dextran sulphate sodium (DSS; 3.5% w/v in drinking water) to evoke intestinal inflammation and tissue damage. CAR-deficient mice were exposed to DSS and mucosal healing assessed. Modulation of wound healing by CAR was assessed in vitro. The therapeutic potential of CAR activation was evaluated, using 3,3',5,5'-tetrachloro-1,4-bis(pyridyloxy)benzene (TCPOBOP), a selective rodent CAR agonist. KEY RESULTS CAR expression was reduced in CD and UC samples, compared with expression in healthy controls. This was reproduced in our DSS studies, where CAR expression was reduced in colitic mice. CAR-deficient mice exhibited reduced healing following DSS exposure. In vitro, CAR activation accelerated intestinal epithelial wound healing by enhancing cell migration. Lastly, treating mice with TCPOBOP, following induction of colitis, enhanced mucosal healing. CONCLUSION AND IMPLICATIONS Our results support the notion that xenobiotic sensing is altered during intestinal inflammation, and suggest that CAR activation may prove effective in enhancing mucosal healing in patients with IBD.
Collapse
Affiliation(s)
- Grace M Hudson
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Canada.,Department of Microbiology, Immunology & Infectious Diseases, University of Calgary, Calgary, Canada
| | - Kyle L Flannigan
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Canada.,Department of Microbiology, Immunology & Infectious Diseases, University of Calgary, Calgary, Canada
| | - Sarah L Erickson
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Canada.,Department of Microbiology, Immunology & Infectious Diseases, University of Calgary, Calgary, Canada
| | - Fernando A Vicentini
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Canada.,Department of Microbiology, Immunology & Infectious Diseases, University of Calgary, Calgary, Canada
| | - Alexandra Zamponi
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Canada.,Department of Microbiology, Immunology & Infectious Diseases, University of Calgary, Calgary, Canada
| | | | - Laurie Alston
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Canada.,Department of Microbiology, Immunology & Infectious Diseases, University of Calgary, Calgary, Canada
| | - Christophe Altier
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Canada
| | - Subrata Ghosh
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Kevin P Rioux
- Department of Medicine, University of Calgary, Calgary, Canada
| | - Sridhar Mani
- Department of Medicine & Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Thomas K Chang
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada
| | - Simon A Hirota
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Canada.,Department of Microbiology, Immunology & Infectious Diseases, University of Calgary, Calgary, Canada
| |
Collapse
|
44
|
Oh-oka K, Kojima Y, Uchida K, Yoda K, Ishimaru K, Nakajima S, Hemmi J, Kano H, Fujii-Kuriyama Y, Katoh R, Ito H, Nakao A. Induction of Colonic Regulatory T Cells by Mesalamine by Activating the Aryl Hydrocarbon Receptor. Cell Mol Gastroenterol Hepatol 2017; 4:135-151. [PMID: 28593185 PMCID: PMC5453907 DOI: 10.1016/j.jcmgh.2017.03.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 03/31/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Mesalamine is a first-line drug for treatment of inflammatory bowel diseases (IBD). However, its mechanisms are not fully understood. CD4+ Foxp3+ regulatory T cells (Tregs) play a potential role in suppressing IBD. This study determined whether the anti-inflammatory activity of mesalamine is related to Treg induction in the colon. METHODS We examined the frequencies of Tregs in the colons of wild-type mice, mice deficient for aryl hydrocarbon receptor (AhR-/- mice), and bone marrow-chimeric mice lacking AhR in hematopoietic cells (BM-AhR-/- mice), following oral treatment with mesalamine. We also examined the effects of mesalamine on transforming growth factor (TGF)-β expression in the colon. RESULTS Treatment of wild-type mice with mesalamine increased the accumulation of Tregs in the colon and up-regulated the AhR target gene Cyp1A1, but this effect was not observed in AhR-/- or BM-AhR-/- mice. In addition, mesalamine promoted in vitro differentiation of naive T cells to Tregs, concomitant with AhR activation. Mice treated with mesalamine exhibited increased levels of the active form of TGF-β in the colon in an AhR-dependent manner and blockade of TGF-β signaling suppressed induction of Tregs by mesalamine in the colon. Furthermore, mice pretreated with mesalamine acquired resistance to dextran sodium sulfate-induced colitis. CONCLUSIONS We propose a novel anti-inflammatory mechanism of mesalamine for colitis: induction of Tregs in the colon via the AhR pathway, followed by TGF-β activation.
Collapse
Key Words
- AhR, aryl hydrocarbon receptor
- Aryl Hydrocarbon Receptor
- BM, bone marrow
- DSS, dextran sodium sulfate
- ELISA, enzyme-linked immunosorbent assay
- FBS, fetal bovine serum
- FITC, fluorescein isothiocyanate
- IBD, inflammatory bowel disease
- IFN, interferon
- IL, interleukin
- LPL, lamina propria lymphocytes
- MLN, mesenteric lymph nodes
- Mesalamine
- PBS, phosphate-buffered saline
- Q-PCR, quantitative polymerase chain reaction
- RPMI, Roswell Park Memorial Institute
- Regulatory T Cells
- TCDD, 2,3,7,8-tetrachlorodibenzo-p-dioxin
- TGF, transforming growth factor
- TGF-β
- TNF, tumor necrosis factor
- Tregs, regulatory T cells
- WT, wild-type
- XRE, xenobiotic responsive element
- mAb, monoclonal antibody
Collapse
Affiliation(s)
- Kyoko Oh-oka
- Department of Immunology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Yuko Kojima
- The Laboratory of Morphology and Image Analysis, Research Support Center, Juntendo University School of Medicine, Tokyo, Japan
| | - Koichiro Uchida
- Atopy Research Center, Juntendo University School of Medicine, Tokyo, Japan
| | - Kimiko Yoda
- Department of Pathology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Kayoko Ishimaru
- Department of Immunology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Shotaro Nakajima
- Department of Immunology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Jun Hemmi
- Food Science Research Laboratories, Division of Research and Development, Meiji Co, Ltd, Kanagawa, Japan
| | - Hiroshi Kano
- Food Science Research Laboratories, Division of Research and Development, Meiji Co, Ltd, Kanagawa, Japan
| | | | - Ryohei Katoh
- Department of Pathology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Hiroyuki Ito
- Food Science Research Laboratories, Division of Research and Development, Meiji Co, Ltd, Kanagawa, Japan
| | - Atsuhito Nakao
- Department of Immunology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan,Atopy Research Center, Juntendo University School of Medicine, Tokyo, Japan,Correspondence Address correspondence to: Atsuhito Nakao, MD, PhD, Department of Immunology, Faculty of Medicine, University of Yamanashi, 1110, Shimokato, Chuo, Yamanashi 409-3898, Japan. fax: 81-55-273-9542.Department of ImmunologyFaculty of MedicineUniversity of Yamanashi1110, ShimokatoChuoYamanashi 409-3898Japan
| |
Collapse
|
45
|
Ding YP, Ladeiro Y, Morilla I, Bouhnik Y, Marah A, Zaag H, Cazals-Hatem D, Seksik P, Daniel F, Hugot JP, Wainrib G, Tréton X, Ogier-Denis E. Integrative Network-based Analysis of Colonic Detoxification Gene Expression in Ulcerative Colitis According to Smoking Status. J Crohns Colitis 2017; 11:474-484. [PMID: 27702825 DOI: 10.1093/ecco-jcc/jjw179] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 10/03/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUNDS AND AIMS The effect of cigarette smoking [CS] is ambivalent since smoking improves ulcerative colitis [UC] while it worsens Crohn's disease [CD]. Although this clinical relationship between inflammatory bowel disease [IBD] and tobacco is well established, only a few experimental works have investigated the effect of smoking on the colonic barrier homeostasis focusing on xenobiotic detoxification genes. METHODS A comprehensive and integrated comparative analysis of the global xenobiotic detoxification capacity of the normal colonic mucosa of healthy smokers [n = 8] and non-smokers [n = 9] versus the non-affected colonic mucosa of UC patients [n = 19] was performed by quantitative real-time polymerase chain reaction [qRT PCR]. The detoxification gene expression profile was analysed in CD patients [n = 18], in smoking UC patients [n = 5], and in biopsies from non-smoking UC patients cultured or not with cigarette smoke extract [n = 8]. RESULTS Of the 244 detoxification genes investigated, 65 were dysregulated in UC patients in comparison with healthy controls or CD patients. The expression of ≥ 45/65 genes was inversed by CS in biopsies of smoking UC patients in remission and in colonic explants of UC patients exposed to cigarette smoke extract. We devised a network-based data analysis approach for differentially assessing changes in genetic interactions, allowing identification of unexpected regulatory detoxification genes that may play a major role in the beneficial effect of smoking on UC. CONCLUSIONS Non-inflamed colonic mucosa in UC is characterised by a specifically altered detoxification gene network, which is partially restored by tobacco. These mucosal signatures could be useful for developing new therapeutic strategies and biomarkers of drug response in UC.
Collapse
Affiliation(s)
- Yong-Ping Ding
- INSERM, Research Centre of Inflammation BP 416, Paris, France.,Université Paris-Diderot Sorbonne Paris-Cité, Paris, France.,Laboratory of Excellence Labex INFLAMEX, Sorbonne-Paris- Cité, Paris, France
| | - Yannick Ladeiro
- INSERM, Research Centre of Inflammation BP 416, Paris, France.,Université Paris-Diderot Sorbonne Paris-Cité, Paris, France.,Laboratory of Excellence Labex INFLAMEX, Sorbonne-Paris- Cité, Paris, France
| | - Ian Morilla
- INSERM, Research Centre of Inflammation BP 416, Paris, France.,Université Paris-Diderot Sorbonne Paris-Cité, Paris, France.,Laboratory of Excellence Labex INFLAMEX, Sorbonne-Paris- Cité, Paris, France.,Université Paris 13, Sorbonne Paris Cité, Villetaneuse, France
| | - Yoram Bouhnik
- INSERM, Research Centre of Inflammation BP 416, Paris, France.,Université Paris-Diderot Sorbonne Paris-Cité, Paris, France.,Laboratory of Excellence Labex INFLAMEX, Sorbonne-Paris- Cité, Paris, France.,Assistance Publique Hôpitaux de Paris, Service de gastroentérologie, MICI et assistance nutritive, Hôpital Beaujon, Clichy la Garenne, France
| | - Assiya Marah
- INSERM, Research Centre of Inflammation BP 416, Paris, France.,Université Paris-Diderot Sorbonne Paris-Cité, Paris, France.,Laboratory of Excellence Labex INFLAMEX, Sorbonne-Paris- Cité, Paris, France
| | - Hatem Zaag
- Laboratory of Excellence Labex INFLAMEX, Sorbonne-Paris- Cité, Paris, France.,Université Paris 13, Sorbonne Paris Cité, Villetaneuse, France
| | - Dominique Cazals-Hatem
- INSERM, Research Centre of Inflammation BP 416, Paris, France.,Université Paris-Diderot Sorbonne Paris-Cité, Paris, France.,Laboratory of Excellence Labex INFLAMEX, Sorbonne-Paris- Cité, Paris, France.,Assistance Publique Hôpitaux de Paris, Service d'anatomopathologie, Hôpital Beaujon, Clichy la Garenne, France
| | - Philippe Seksik
- INSERM U1157, UMR 7203, F-7502, Paris, France.,Assistance Publique Hôpitaux de Paris, Hôpital Saint-Antoine, Paris, France
| | - Fanny Daniel
- INSERM, Research Centre of Inflammation BP 416, Paris, France.,Université Paris-Diderot Sorbonne Paris-Cité, Paris, France.,Laboratory of Excellence Labex INFLAMEX, Sorbonne-Paris- Cité, Paris, France
| | - Jean-Pierre Hugot
- INSERM, Research Centre of Inflammation BP 416, Paris, France.,Université Paris-Diderot Sorbonne Paris-Cité, Paris, France.,Laboratory of Excellence Labex INFLAMEX, Sorbonne-Paris- Cité, Paris, France.,Assistance Publique Hôpitaux de Paris, Hôpital Robert Debré, Paris, France
| | - Gilles Wainrib
- Laboratory of Excellence Labex INFLAMEX, Sorbonne-Paris- Cité, Paris, France.,Département d'Informatique, Equipe DATA, Ecole Normale Supérieure, Paris, France
| | - Xavier Tréton
- INSERM, Research Centre of Inflammation BP 416, Paris, France.,Université Paris-Diderot Sorbonne Paris-Cité, Paris, France.,Laboratory of Excellence Labex INFLAMEX, Sorbonne-Paris- Cité, Paris, France.,Assistance Publique Hôpitaux de Paris, Service de gastroentérologie, MICI et assistance nutritive, Hôpital Beaujon, Clichy la Garenne, France
| | - Eric Ogier-Denis
- INSERM, Research Centre of Inflammation BP 416, Paris, France.,Université Paris-Diderot Sorbonne Paris-Cité, Paris, France.,Laboratory of Excellence Labex INFLAMEX, Sorbonne-Paris- Cité, Paris, France
| |
Collapse
|
46
|
Islam J, Sato S, Watanabe K, Watanabe T, Ardiansyah, Hirahara K, Aoyama Y, Tomita S, Aso H, Komai M, Shirakawa H. Dietary tryptophan alleviates dextran sodium sulfate-induced colitis through aryl hydrocarbon receptor in mice. J Nutr Biochem 2017; 42:43-50. [PMID: 28113104 DOI: 10.1016/j.jnutbio.2016.12.019] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 12/27/2016] [Accepted: 12/28/2016] [Indexed: 12/15/2022]
Abstract
Ulcerative colitis is the typical progression of chronic inflammatory bowel disease. Amino acids, particularly tryptophan, have been reported to exert a protective effect against colitis induced by dextran sodium sulfate (DSS), but the precise underlying mechanisms remain incompletely clarified. Tryptophan metabolites are recognized to function as endogenous ligands for aryl hydrocarbon receptor (Ahr), which is a critical regulator of inflammation and immunity. Thus, we conducted this study to investigate whether dietary tryptophan supplementation protects against DSS-induced colitis by acting through Ahr. Female wild-type (WT) and Ahr-deficient (knockout; KO) mice (10-12 weeks old) were divided into four groups and fed either a control or 0.5% tryptophan diet. The tryptophan diet ameliorated DSS-induced colitis symptoms and severity in WT mice but not in KO mice, and the diet reduced the mRNA expression of Il-6, Tnfα, Il-1β and the chemokines Ccl2, Cxcl1 and Cxcl2 in the WT groups. Furthermore, Il-22 and Stat3 mRNA expression in the colon was elevated in WT mice fed with the tryptophan diet, which mainly protected epithelial layer integrity, and Ahr also modulated immune homeostasis by regulating Foxp3 and Il-17 mRNA expression. These data suggest that tryptophan-containing diet might ameliorate DSS-induced acute colitis and regulate epithelial homeostasis through Ahr. Thus, tryptophan could serve as a promising preventive agent in the treatment of ulcerative colitis.
Collapse
Affiliation(s)
- Jahidul Islam
- Laboratory of Nutrition, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Shoko Sato
- Department of Biological Science and Technology, Tokyo University of Science, Tokyo, Japan
| | - Kouichi Watanabe
- Cellular Biology Laboratory, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan; International Education and Research Center for Food Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Takaya Watanabe
- Laboratory of Nutrition, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Ardiansyah
- Laboratory of Nutrition, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan; Department of Food Science and Technology, Universitas Bakrie, Jakarta, Indonesia
| | - Keisuke Hirahara
- Laboratory of Nutrition, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Yukihide Aoyama
- Laboratory of Nutrition, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Shuhei Tomita
- Department of Pharmacology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Hisashi Aso
- Cellular Biology Laboratory, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan; International Education and Research Center for Food Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Michio Komai
- Laboratory of Nutrition, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Hitoshi Shirakawa
- Laboratory of Nutrition, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan; International Education and Research Center for Food Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.
| |
Collapse
|
47
|
Vogel CFA, Haarmann-Stemmann T. The aryl hydrocarbon receptor repressor - More than a simple feedback inhibitor of AhR signaling: Clues for its role in inflammation and cancer. CURRENT OPINION IN TOXICOLOGY 2017; 2:109-119. [PMID: 28971163 DOI: 10.1016/j.cotox.2017.02.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The aryl hydrocarbon receptor repressor (AhRR) was first described as a specific competitive repressor of aryl hydrocarbon receptor (AhR) activity based on its ability to dimerize with the AhR nuclear translocator (ARNT) and through direct competition of AhR/ARNT and AhRR/ARNT complexes for binding to dioxin-responsive elements (DREs). Like AhR, AhRR belongs to the basic Helix-Loop-Helix/Per-ARNT-Sim (bHLH/PAS) protein family but lacks functional ligand-binding and transactivation domains. Transient transfection experiments with ARNT and AhRR mutants examining the inhibitory mechanism of AhRR suggested a more complex mechanism than the simple mechanism of negative feedback through sequestration of ARNT to regulate AhR signaling. Recently, AhRR has been shown to act as a tumor suppressor gene in several types of cancer cells. Furthermore, epidemiological studies have found epigenetic changes and silencing of AhRR associated with exposure to cigarette smoke and cancer development. Additional studies from our laboratories have demonstrated that AhRR represses other signaling pathways including NF-κB and is capable of regulating inflammatory responses. A better understanding of the regulatory mechanisms of AhRR in AhR signaling and adverse outcome pathways leading to deregulated inflammatory responses contributing to tumor promotion and other adverse health effects is expected from future studies. This review article summarizes the characteristics of AhRR as an inhibitor of AhR activity and highlights more recent findings pointing out the role of AhRR in inflammation and tumorigenesis.
Collapse
Affiliation(s)
- Christoph F A Vogel
- Department of Environmental Toxicology and Center for Health and the Environment, University of California, Davis, CA 95616, USA
| | | |
Collapse
|
48
|
Ehrlich AK, Kerkvliet NI. Is chronic AhR activation by rapidly metabolized ligands safe for the treatment of immune-mediated diseases? CURRENT OPINION IN TOXICOLOGY 2017; 2:72-78. [PMID: 28944315 DOI: 10.1016/j.cotox.2017.01.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
There is a long standing perception that AhR ligands are automatically disqualified from pharmaceutical development due to their induction of Cyp1a1 as well as their potential for causing "dioxin-like" toxicities. However, recent discoveries of new AhR ligands with potential therapeutic applications have been reported, inviting reconsideration of this policy. One area of exploration is focused on the activation of AhR to promote the generation of regulatory T cells, which control the intensity and duration of immune responses. Rapidly metabolized AhR ligands (RMAhRLs), which do not bioaccumulate in the same manner as 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) have been discovered that induce Tregs and display impressive therapeutic efficacy in a broad range of preclinical models of immune-mediated diseases. Given the promise of these RMAhRLs, is the bias against AhR activators still valid? Can RMAhRLs be given chronically to maintain therapeutic levels of AhR activation without producing the same toxicity profile as dioxin-like compounds? Based on our review of the data, there is little evidence to support the indiscriminate exclusion of AhR activators/Cyp1a1 inducers from early drug developmental pipelines. We also found no evidence that short-term treatment with RMAhRLs produce "dioxin-like toxicity" and, in fact, were well tolerated. However, safety testing of individual RMAhRLs under therapeutic conditions, as performed with all promising new drugs, will be needed to reveal whether or not chronic activation of AhR leads to unacceptable adverse outcomes.
Collapse
Affiliation(s)
- Allison K Ehrlich
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, 97331, United States
| | - Nancy I Kerkvliet
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, 97331, United States
| |
Collapse
|
49
|
Murray IA, Perdew GH. Ligand activation of the Ah receptor contributes to gastrointestinal homeostasis. CURRENT OPINION IN TOXICOLOGY 2017; 2:15-23. [PMID: 28944314 DOI: 10.1016/j.cotox.2017.01.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The Ah receptor (AHR) is capable of binding a structurally diverse group of compounds that can be found in the diet, produced by bacteria in the gut and through endogenous metabolism. The gastrointestinal tract is a rich source of AHR ligands, which have been shown to protect the gut upon challenge with either pathogenic bacteria or toxic chemicals. The human AHR can be activated by a broader range of ligands compared to the mouse AHR, suggesting that studies in mice may underestimate the impact of AHR ligands in the human gut. The protective effect of AHR activation appears to be due to modulating the immune system within the gut. While several mechanisms have been established, due to the increasingly pleotropic nature of the AHR, other mechanisms of action likely exist that remain to be identified. The major contributors to AHR function in the gut and the most appropriate level of receptor activation that maintains intestinal homeostasis warrants further investigation.
Collapse
Affiliation(s)
- Iain A Murray
- Department of Veterinary and Biomedical Sciences, and Center for Molecular Toxicology and Carcinogenesis, Penn State University, University Park, PA 16802
| | - Gary H Perdew
- Department of Veterinary and Biomedical Sciences, and Center for Molecular Toxicology and Carcinogenesis, Penn State University, University Park, PA 16802
| |
Collapse
|
50
|
Goettel JA, Gandhi R, Kenison JE, Yeste A, Murugaiyan G, Sambanthamoorthy S, Griffith AE, Patel B, Shouval DS, Weiner HL, Snapper SB, Quintana FJ. AHR Activation Is Protective against Colitis Driven by T Cells in Humanized Mice. Cell Rep 2016; 17:1318-1329. [PMID: 27783946 PMCID: PMC5106873 DOI: 10.1016/j.celrep.2016.09.082] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 08/14/2016] [Accepted: 09/23/2016] [Indexed: 02/07/2023] Open
Abstract
Existing therapies for inflammatory bowel disease that are based on broad suppression of inflammation result in variable clinical benefit and unwanted side effects. A potential therapeutic approach for promoting immune tolerance is the in vivo induction of regulatory T cells (Tregs). Here we report that activation of the aryl hydrocarbon receptor using the non-toxic agonist 2-(1'H-indole-3'-carbonyl)-thiazole-4-carboxylic acid methyl ester (ITE) induces human Tregs in vitro that suppress effector T cells through a mechanism mediated by CD39 and Granzyme B. We then developed a humanized murine system whereby human CD4+ T cells drive colitis upon exposure to 2,4,6-trinitrobenzenesulfonic acid and assessed ITE as a potential therapeutic. ITE administration ameliorated colitis in humanized mice with increased CD39, Granzyme B, and IL10-secreting human Tregs. These results develop an experimental model to investigate human CD4+ T responses in vivo and identify the non-toxic AHR agonist ITE as a potential therapy for promoting immune tolerance in the intestine.
Collapse
Affiliation(s)
- Jeremy A Goettel
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Roopali Gandhi
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Neurology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Jessica E Kenison
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Neurology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Ada Yeste
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Neurology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Gopal Murugaiyan
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Neurology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Sharmila Sambanthamoorthy
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Neurology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Alexandra E Griffith
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA 02115, USA
| | - Bonny Patel
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Neurology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Dror S Shouval
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Neurology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Scott B Snapper
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA 02115, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Division of Gastroenterology, Brigham and Women's Hospital, Boston, MA 02115, USA.
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Neurology, Brigham and Women's Hospital, Boston, MA 02115, USA; The Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA.
| |
Collapse
|