1
|
Huang Y, Xu W, Dong W, Chen G, Sun Y, Zeng X. Anti-diabetic effect of dicaffeoylquinic acids is associated with the modulation of gut microbiota and bile acid metabolism. J Adv Res 2025; 72:17-35. [PMID: 38969095 DOI: 10.1016/j.jare.2024.06.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 06/06/2024] [Accepted: 06/30/2024] [Indexed: 07/07/2024] Open
Abstract
INTRODUCTION The human gut microbiome plays a pivotal role in health and disease, notably through its interaction with bile acids (BAs). BAs, synthesized in the liver, undergo transformation by the gut microbiota upon excretion into the intestine, thus influencing host metabolism. However, the potential mechanisms of dicaffeoylquinic acids (DiCQAs) from Ilex kudingcha how to modulate lipid metabolism and inflammation via gut microbiota remain unclear. OBJECTIVES AND METHODS The objectives of the present study were to investigate the regulating effects of DiCQAs on diabetes and the potential mechanisms of action. Two mice models were utilized to investigate the anti-diabetic effects of DiCQAs. Additionally, analysis of gut microbiota structure and functions was conducted concurrently with the examination of DiCQAs' impact on gut microbiota carrying the bile salt hydrolase (BSH) gene, as well as on the enterohepatic circulation of BAs and related signaling pathways. RESULTS Our findings demonstrated that DiCQAs alleviated diabetic symptoms by modulating gut microbiota carrying the BSH gene. This modulation enhanced intestinal barrier integrity, increased enterohepatic circulation of conjugated BAs, and inhibited the farnesoid X receptor-fibroblast growth factor 15 (FGF15) signaling axis in the ileum. Consequently, the protein expression of hepatic FGFR4 fibroblast growth factor receptor 4 (FGFR4) decreased, accompanied by heightened BA synthesis, reduced hepatic BA stasis, and lowered levels of hepatic and plasma cholesterol. Furthermore, DiCQAs upregulated glucolipid metabolism-related proteins in the liver and muscle, including v-akt murine thymoma viral oncogene homolog (AKT)/glycogen synthase kinase 3-beta (GSK3β) and AMP-activated protein kinase (AMPK), thereby ameliorating hyperglycemia and mitigating inflammation through the down-regulation of the MAPK signaling pathway in the diabetic group. CONCLUSION Our study elucidated the anti-diabetic effects and mechanism of DiCQAs from I. kudingcha, highlighting the potential of targeting gut microbiota, particularly Acetatifactor sp011959105 and Acetatifactor muris carrying the BSH gene, as a therapeutic strategy to attenuate FXR-FGF15 signaling and ameliorate diabetes.
Collapse
Affiliation(s)
- Yujie Huang
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China; School of Public Health, Guizhou Medical University, Guiyang 561113, Guizhou, China
| | - Weiqi Xu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Wei Dong
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Guijie Chen
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Yi Sun
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Xiaoxiong Zeng
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China.
| |
Collapse
|
2
|
Yao L, Zhou X, Jiang X, Chen H, Li Y, Xiong X, Tang Y, Zhang H, Qiao P. High-fat diet promotes gestational diabetes mellitus through modulating gut microbiota and bile acid metabolism. Front Microbiol 2025; 15:1480446. [PMID: 39935515 PMCID: PMC11810896 DOI: 10.3389/fmicb.2024.1480446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 12/27/2024] [Indexed: 02/13/2025] Open
Abstract
Introduction Gestational diabetes mellitus (GDM) is a condition characterized by glucose intolerance during pregnancy, estimated to affect approximately 20% of the whole pregnancies and is increasing in prevalence globally. However, there is still a big gap in knowledge about the association between gut microbiota associated metabolism alterations and GDM development. Methods All the participants accomplished the validated internet-based dietary questionnaire for Chinese and serum, fecal samples were collected. HFD, control diet or colesevelam intervention was fed to GDM mice models or Fxr-/- mice models, with or without antibiotics cocktail treatment. Fecal microbiota transplantation were used for further validation. Gut microbiota and metabolites were detected by metagenomic sequencing and high-performance liquid chromatography-mass spectrometry, respectively. Bile acids of serum, fecal samples from human and mice were analysised. Body weight, average feed intake, blood glucose, insulin levels and oral glucose tolerance test was performed among each groups. Expression levels of Fxr, Shp and Fgf15 mRNA and protein were detected by quantitative reverse transcription polymerase chain reaction and western blot, respectively. Results Our data indicated that high fat diet (HFD) was linked with higher prevalence of GDM, and HFD was positively associated with poor prognosis in GDM patients. Moreover, compared with normal diet (ND) group, GDM patients from HFD group performed a loss of gut microbiota diversity and enrichment of Alistipes onderdonkii, Lachnospiraceae bacterium 1_7_58FAA, and Clostridium aspaaragiforme while ruduction of Akkermansiaceae, Paraprevotell xylaniphila, and Prevotella copri. Additionally, HFD aggravated GDM in mice and gut microbiota depletion by antibiotics crippled the effect of excess fat intake. BAs profile altered in HFD GDM patients and mice models. Fecal microbiota transplantation (FMT) further confirmed that gut microbiota contributed to bile acids (BAs) metabolic dysfunction during HFD-associated GDM development. Mechanically, HFD-FMT administration activated Fxr, Shp, and Fgf15 activity, disturbed the glucose metabolism and aggravated insulin resistance but not in HFD-FMT Fxr-/- mice and ND-FMT Fxr-/- mice. Furthermore, colesevelam intervention alleviated HFD-associated GDM development, improved BAs metabolism, suppressed Fxr, Shp, and Fgf15 activity only in WT mice but not in the Fxr-/- HFD + Colesevelam group and Fxr-/- HFD group. HFD induced GDM and contributed to poor prognosis in GDM parturients through inducing gut microbial dysbiosis and metabolic alteration, especially appeared in BAs profile. Moreover, Fxr pathway participated in regulating HFD-associated gut microbiota disordered BAs metabolites and aggravating GDM in mice. Discussion Modulating gut microbiota and BAs metabolites could be a potential therapeutic strategy in the prevention and treatment of HFD-associated GDM.
Collapse
Affiliation(s)
- Lei Yao
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xuefei Zhou
- Department of Gynaecology and Obstetrics, The Center of Red Cross Hospital of Harbin, Harbin, China
| | - Xianqi Jiang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hao Chen
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuanliang Li
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiao Xiong
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yan Tang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Haogang Zhang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Pengfei Qiao
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
3
|
Geng X, Zhuang M, Tian W, Shang H, Gong Z, Lv Y, Li J. Green Radish Polysaccharide Prevents Alcoholic Liver Injury by Interfering with Intestinal Bacteria and Short-Chain Fatty Acids in Mice. Foods 2024; 13:3733. [PMID: 39682806 DOI: 10.3390/foods13233733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
This study aimed to ascertain the potential benefits of green radish polysaccharide (GRP) in treating alcoholic liver disease (ALD) in mice and explore its mechanism of action. Using biochemical analysis, high-throughput sequencing of gut microbiota, and gas chromatography-mass spectrometry to measure short-chain fatty acids (SCFAs) in feces, we found that GRP intervention significantly improved lipid metabolism and hepatic function in mice subjected to excessive alcohol intake. The GRP intervention reduced malondialdehyde levels by 66% and increased total superoxide dismutase levels by 22%, thereby mitigating alcohol-induced oxidative stress. Furthermore, GRP intervention in mice with alcohol consumption resulted in a reduction in tumor necrosis factor, interleukin 6, and lipopolysaccharide levels by 12%, 9%, and 25%, respectively, effectively attenuating alcoholic liver inflammation. 16S rRNA amplicon sequencing demonstrated that excessive alcohol consumption markedly altered the gut microbiota composition in mice. The GRP treatment resulted in a significant reduction in the number of beneficial bacteria (Lactobacillus and Lachnospiraceae_NK4A136_group) and an increase in the proportion of harmful bacteria (Muribaculaceae and Verrucomicrobiota). The metabolomic analyses of the SCFAs demonstrated an increase in the contents of SCFAs, acetic acid, propionic acid, and butyric acid, following GRP supplementation. Furthermore, the metabolic levels of cholinergic synapses and glycolysis/gluconeogenesis were found to be modulated. In conclusion, these findings suggest that GRP may attenuate alcohol-induced oxidative damage in the liver by modulating the gut microbiota and hepatic metabolic pathways. This may position GRP as a potential functional component for ALD prevention.
Collapse
Affiliation(s)
- Xiong Geng
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| | - Miaomiao Zhuang
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| | - Weina Tian
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| | - Huayan Shang
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| | - Ziyi Gong
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| | - Yanfang Lv
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| | - Jianrong Li
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| |
Collapse
|
4
|
Sun L, Jiang J, Zeng Y, Zhu J, Wang S, Huang D, Cao C. Polysaccharide NAP-3 Synergistically Enhances the Efficiency of Metformin in Type 2 Diabetes via Bile Acid/GLP-1 Axis through Gut Microbiota Remodeling. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:21077-21088. [PMID: 39262139 DOI: 10.1021/acs.jafc.4c06030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
The polysaccharides of edible mushrooms are excellent phytochemicals for adjuvant treatment of metabolic diseases, but the potential mechanisms of synergistic effects are unclear. In this work, we discovered that NAP-3 enhanced the efficiency of metformin in lipid and glucose metabolism in type 2 diabetic (T2D) mice in a gut microbiome-dependent way. NAP-3 remodeled the intestinal microbial, resulting in the decreased activity of bile salt hydrolases and upregulation of CYP27A1 and CYP7B1 functions in the alternative pathway of bile acid synthesis, which leads to accumulation of the conjugated bile acids in ileum, specifically TβMCA and TUDCA. The accumulated conjugated bile acids either blocked or stimulated the nuclear receptors Farnesoid-X-receptor and TGR5, inducing the release of GLP-1 and ultimately enhanced glucose metabolism in mice. Collectively, our research indicated that edible mushroom polysaccharide NAP-3 may serve as a promising adjunctive oral therapeutic agent for T2D.
Collapse
Affiliation(s)
- Lu Sun
- Department of Food Quality and Safety/National R&D Center for Chinese Herbal Medicine Processing, College of Engineering, China Pharmaceutical University, 211198 Nanjing, China
| | - Jiang Jiang
- Department of Food Quality and Safety/National R&D Center for Chinese Herbal Medicine Processing, College of Engineering, China Pharmaceutical University, 211198 Nanjing, China
| | - Yan Zeng
- Department of Food Quality and Safety/National R&D Center for Chinese Herbal Medicine Processing, College of Engineering, China Pharmaceutical University, 211198 Nanjing, China
| | - Jie Zhu
- Department of Food Quality and Safety/National R&D Center for Chinese Herbal Medicine Processing, College of Engineering, China Pharmaceutical University, 211198 Nanjing, China
| | - Suilou Wang
- Department of Food Quality and Safety/National R&D Center for Chinese Herbal Medicine Processing, College of Engineering, China Pharmaceutical University, 211198 Nanjing, China
| | - Dechun Huang
- Department of Food Quality and Safety/National R&D Center for Chinese Herbal Medicine Processing, College of Engineering, China Pharmaceutical University, 211198 Nanjing, China
| | - Chongjiang Cao
- Department of Food Quality and Safety/National R&D Center for Chinese Herbal Medicine Processing, College of Engineering, China Pharmaceutical University, 211198 Nanjing, China
| |
Collapse
|
5
|
Beekman CN, Penumutchu S, Peterson R, Han G, Belenky M, Hasan MH, Belenky A, Beura LK, Belenky P. Spatial analysis of murine microbiota and bile acid metabolism during amoxicillin treatment. Cell Rep 2024; 43:114572. [PMID: 39116202 DOI: 10.1016/j.celrep.2024.114572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 06/05/2024] [Accepted: 07/17/2024] [Indexed: 08/10/2024] Open
Abstract
Antibiotics cause collateral damage to resident microbes that is associated with various health risks. To date, studies have largely focused on the impacts of antibiotics on large intestinal and fecal microbiota. Here, we employ a gastrointestinal (GI) tract-wide integrated multiomic approach to show that amoxicillin (AMX) treatment reduces bacterial abundance, bile salt hydrolase activity, and unconjugated bile acids in the small intestine (SI). Losses of fatty acids (FAs) and increases in acylcarnitines in the large intestine (LI) correspond with spatially distinct expansions of Proteobacteria. Parasutterella excrementihominis engage in FA biosynthesis in the SI, while multiple Klebsiella species employ FA oxidation during expansion in the LI. We subsequently demonstrate that restoration of unconjugated bile acids can mitigate losses of commensals in the LI while also inhibiting the expansion of Proteobacteria during AMX treatment. These results suggest that the depletion of bile acids and lipids may contribute to AMX-induced dysbiosis in the lower GI tract.
Collapse
Affiliation(s)
- Chapman N Beekman
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA.
| | - Swathi Penumutchu
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Rachel Peterson
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Geongoo Han
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Marina Belenky
- Felicitex Therapeutics Inc., 27 Strathmore Road, Natick, MA 01760, USA
| | - Mohammad H Hasan
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Alexei Belenky
- Felicitex Therapeutics Inc., 27 Strathmore Road, Natick, MA 01760, USA
| | - Lalit K Beura
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Peter Belenky
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
6
|
Sheng X, Zhan P, Wang P, He W, Tian H. Mitigation of high-fat diet-induced hepatic steatosis by thyme ( Thymus quinquecostatus Celak) polyphenol-rich extract (TPE): insights into gut microbiota modulation and bile acid metabolism. Food Funct 2024; 15:7333-7347. [PMID: 38305590 DOI: 10.1039/d3fo05235d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
Our previous study demonstrated that thyme polyphenol-rich extract (TPE) mitigated hepatic injury induced by a high-fat diet (HFD) through the regulation of lipid metabolism, promotion of short-chain fatty acid production, enhancement of intestinal barrier function, and attenuation of inflammation. In this study, we aimed to further elucidate additional mechanisms underlying TPE-mediated preventive effects on hepatic steatosis, with a specific focus on its impact on the gut microbiota and bile acid (BA) metabolism in HFD-fed mice. TPE treatment resulted in a significant reduction in serum total BA levels and a notable increase in fecal total BA levels. In particular, elevations in fecal conjugated BA levels, in turn, impede intestinal farnesoid X receptor (FXR) signaling, thereby enhancing hepatic synthesis and fecal excretion of BAs. The downregulated mRNA expression levels of intestinal Fxr and Fgf15, and hepatic Fgfr4, along with the upregulated mRNA expression levels of Cyp7a1 and Cyp27a1 after TPE treatment also prove the above inference. Meanwhile, TPE appeared to promote BA efflux and enterohepatic circulation, as evidenced by changes in the mRNA levels of Bsep, Ntpc, Shp, Asbt, Ibabp, and Ostα/β. TPE also modulated the gut microbiota and was characterized by an increased relative abundance of Lactobacillus. Furthermore, antibiotic treatment depleted the intestinal flora in mice, also abrogating the hepatoprotective effect of TPE against NAFLD. These findings collectively indicate that TPE effectively mitigates HFD-induced NAFLD by modulating the gut-liver axis, specifically targeting the gut microbiota and bile acid metabolism.
Collapse
Affiliation(s)
- Xialu Sheng
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China.
| | - Ping Zhan
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China.
| | - Peng Wang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China.
| | - Wanying He
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China.
| | - Honglei Tian
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
7
|
Yang M, Zheng X, Fan J, Cheng W, Yan T, Lai Y, Zhang N, Lu Y, Qi J, Huo Z, Xu Z, Huang J, Jiao Y, Liu B, Pang R, Zhong X, Huang S, Luo G, Lee G, Jobin C, Eren AM, Chang EB, Wei H, Pan T, Wang X. Antibiotic-Induced Gut Microbiota Dysbiosis Modulates Host Transcriptome and m 6A Epitranscriptome via Bile Acid Metabolism. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307981. [PMID: 38713722 PMCID: PMC11267274 DOI: 10.1002/advs.202307981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 04/15/2024] [Indexed: 05/09/2024]
Abstract
Gut microbiota can influence host gene expression and physiology through metabolites. Besides, the presence or absence of gut microbiome can reprogram host transcriptome and epitranscriptome as represented by N6-methyladenosine (m6A), the most abundant mammalian mRNA modification. However, which and how gut microbiota-derived metabolites reprogram host transcriptome and m6A epitranscriptome remain poorly understood. Here, investigation is conducted into how gut microbiota-derived metabolites impact host transcriptome and m6A epitranscriptome using multiple mouse models and multi-omics approaches. Various antibiotics-induced dysbiotic mice are established, followed by fecal microbiota transplantation (FMT) into germ-free mice, and the results show that bile acid metabolism is significantly altered along with the abundance change in bile acid-producing microbiota. Unbalanced gut microbiota and bile acids drastically change the host transcriptome and the m6A epitranscriptome in multiple tissues. Mechanistically, the expression of m6A writer proteins is regulated in animals treated with antibiotics and in cultured cells treated with bile acids, indicating a direct link between bile acid metabolism and m6A biology. Collectively, these results demonstrate that antibiotic-induced gut dysbiosis regulates the landscape of host transcriptome and m6A epitranscriptome via bile acid metabolism pathway. This work provides novel insights into the interplay between microbial metabolites and host gene expression.
Collapse
Affiliation(s)
- Meng Yang
- School of Life SciencesSouth China Normal UniversityGuangzhou510631China
| | - Xiaoqi Zheng
- School of Life SciencesSouth China Normal UniversityGuangzhou510631China
- Guangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhou510530China
| | - Jiajun Fan
- School of Life SciencesSouth China Normal UniversityGuangzhou510631China
| | - Wei Cheng
- College of Animal Science and TechnologyHuazhong Agricultural UniversityWuhan430070China
| | - Tong‐Meng Yan
- State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyTaipaMacau999078China
| | - Yushan Lai
- School of Life SciencesSouth China Normal UniversityGuangzhou510631China
| | - Nianping Zhang
- School of Life SciencesSouth China Normal UniversityGuangzhou510631China
| | - Yi Lu
- School of Life SciencesSouth China Normal UniversityGuangzhou510631China
- Guangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhou510530China
| | - Jiali Qi
- School of Life SciencesSouth China Normal UniversityGuangzhou510631China
| | - Zhengyi Huo
- School of Life SciencesSouth China Normal UniversityGuangzhou510631China
| | - Zihe Xu
- School of Life SciencesSouth China Normal UniversityGuangzhou510631China
- Guangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhou510530China
| | - Jia Huang
- School of Life SciencesSouth China Normal UniversityGuangzhou510631China
| | - Yuting Jiao
- School of Life SciencesSouth China Normal UniversityGuangzhou510631China
| | - Biaodi Liu
- MOE Key Laboratory of Gene Function and RegulationState Key Laboratory of BiocontrolSchool of Life SciencesSun Yat‐sen UniversityGuangzhou510275China
| | - Rui Pang
- Guangdong Provincial Key Laboratory of Microbial Safety and HealthState Key Laboratory of Applied Microbiology Southern ChinaInstitute of MicrobiologyGuangdong Academy of SciencesGuangzhou510070China
| | - Xiang Zhong
- College of Animal Science and TechnologyNanjing Agricultural UniversityNanjing210095China
| | - Shi Huang
- Faculty of DentistryThe University of Hong KongHong Kong SARChina
| | - Guan‐Zheng Luo
- MOE Key Laboratory of Gene Function and RegulationState Key Laboratory of BiocontrolSchool of Life SciencesSun Yat‐sen UniversityGuangzhou510275China
| | - Gina Lee
- Department of Microbiology and Molecular GeneticsChao Family Comprehensive Cancer CenterUniversity of California Irvine School of MedicineIrvineCA92697USA
| | - Christian Jobin
- Department of MedicineUniversity of Florida College of MedicineGainesvilleFL32610USA
| | - A. Murat Eren
- Helmholtz Institute for Functional Marine Biodiversity26129OldenburgGermany
- Institute for Chemistry and Biology of the Marine EnvironmentUniversity of Oldenburg26129OldenburgGermany
| | - Eugene B Chang
- Department of MedicineKnapp Center for Biomedical DiscoveryThe University of Chicago Knapp Center for Biomedical DiscoveryChicagoIL60637USA
| | - Hong Wei
- College of Animal Science and TechnologyHuazhong Agricultural UniversityWuhan430070China
| | - Tao Pan
- Department of Biochemistry and Molecular BiologyThe University of ChicagoChicagoIL60637USA
| | - Xiaoyun Wang
- School of Life SciencesSouth China Normal UniversityGuangzhou510631China
- Guangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhou510530China
- University of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
8
|
Wang P, Sun J, Zhao W, Wang D, Ma Y, Zhao Y, Wang Y, Zhao X. Tomato Pectin Ameliorated Hepatic Steatosis in High-Fat-Diet Mice by Modulating Gut Microbiota and Bile Acid Metabolism. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024. [PMID: 38856079 DOI: 10.1021/acs.jafc.4c01598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a worldwide public health issue. Changes in the gut microbiota structure and composition are closely related to host pathophysiology processes. Pectin is associated with several beneficial health effects. In the present study, we aimed at investigating the effect of tomato pectin (TP) on hepatic steatosis and exploring the underlying mechanisms by focusing on the regulation of the gut microbiota-bile acid axis. Our results showed that TP attenuated high-fat diet (HFD)-induced liver steatosis and inflammation. TP administration increased the diversity of gut microbiota, enhancing the abundance of beneficial bacteria and suppressing the abundance of harmful or conditional pathogenic bacteria. Further antibiotic-caused microbiome depletion confirmed that the anti-NAFLD activities of TP were dependent on the regulation of gut microbiota. Besides, TP intervention affected feces bile acid metabolism and caused significant changes in functional conjugated bile acids, which in turn inhibited the ileum FXR/FGF15 signaling, leading to stimulation of the hepatic bile acid (BA) production. Furthermore, TP treatment accelerated BA excretion, promoted BA transportation, inhibited BA reabsorption, and facilitated cholesterol efflux to relieve HFD-induced hyperlipidemia. These findings provide a potential dietary intervention strategy for TP against NAFLD via modulation of cross-talk between BAs and gut bacteria.
Collapse
Affiliation(s)
- Pan Wang
- Institute of Agri-food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing 100097, China
| | - Jing Sun
- Institute of Agri-food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing 100097, China
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110866, China
| | - Wenting Zhao
- Institute of Agri-food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing 100097, China
| | - Dan Wang
- Institute of Agri-food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing 100097, China
| | - Yue Ma
- Institute of Agri-food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing 100097, China
| | - Yuanyuan Zhao
- Institute of Agri-food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing 100097, China
| | - Yubin Wang
- Institute of Agri-food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing 100097, China
| | - Xiaoyan Zhao
- Institute of Agri-food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing 100097, China
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110866, China
| |
Collapse
|
9
|
Kawana T, Imoto H, Tanaka N, Tsuchiya T, Yamamura A, Saijo F, Maekawa M, Tamahara T, Shimizu R, Nakagawa K, Ohnuma S, Kamei T, Unno M. The Significance of Bile in the Biliopancreatic Limb on Metabolic Improvement After Duodenal-Jejunal Bypass. Obes Surg 2024; 34:1665-1673. [PMID: 38512643 PMCID: PMC11031486 DOI: 10.1007/s11695-024-07176-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 03/23/2024]
Abstract
INTRODUCTION Duodenal-jejunal bypass (DJB) is an experimental procedure in metabolic surgery that does not have a restrictive component. Changes in bile acid (BA) dynamics and intestinal microbiota are possibly related to metabolic improvement after DJB. Our previous studies involving obese diabetic rats showed the crucial role of the biliopancreatic limb (BPL) in metabolic improvement after DJB caused by BA reabsorption. We established a new DJB procedure to prevent bile from flowing into the BPL and aimed to elucidate the importance of bile in the BPL after DJB. METHODS Otsuka Long-Evans Tokushima Fatty rats with diabetes were divided into three groups: two DJB groups and a sham group (n = 11). Duodenal-jejunal anastomosis was performed proximal to the papilla of Vater in the DJB group (n = 11). However, the DJB-D group (n = 11) underwent a new procedure with duodenal-jejunal anastomosis distal to the papilla of Vater for preventing bile flow into the BPL. RESULTS Glucose metabolism improved and weight gain was suppressed in the DJB group, but not in the DJB-D and sham groups. Serum BA level and conjugated BA concentration were elevated in the DJB group. The gut microbiota was altered only in the DJB group; the abundance of Firmicutes and Bacteroidetes decreased and that of Actinobacteria increased. However, the DJB-D group exhibited no apparent change in the gut microbiota, similar to the sham group. CONCLUSION BAs are essential in the BPL for metabolic improvement after DJB; they can improve the gut microbiota in these processes.
Collapse
Affiliation(s)
- Tomomi Kawana
- Department of Surgery, Tohoku University Graduate School of Medicine, 1-1, Seiryo-Machi, Aoba-Ku, Sendai, 980-8574, Japan
| | - Hirofumi Imoto
- Department of Surgery, Tohoku University Graduate School of Medicine, 1-1, Seiryo-Machi, Aoba-Ku, Sendai, 980-8574, Japan.
| | - Naoki Tanaka
- Department of Surgery, Tohoku University Graduate School of Medicine, 1-1, Seiryo-Machi, Aoba-Ku, Sendai, 980-8574, Japan
| | - Takahiro Tsuchiya
- Department of Surgery, Tohoku University Graduate School of Medicine, 1-1, Seiryo-Machi, Aoba-Ku, Sendai, 980-8574, Japan
| | - Akihiro Yamamura
- Department of Surgery, Tohoku University Graduate School of Medicine, 1-1, Seiryo-Machi, Aoba-Ku, Sendai, 980-8574, Japan
| | - Fumito Saijo
- Department of Surgery, Tohoku University Graduate School of Medicine, 1-1, Seiryo-Machi, Aoba-Ku, Sendai, 980-8574, Japan
| | - Masamitsu Maekawa
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan
| | - Toru Tamahara
- Tohoku University, Tohoku Medical Megabank Organization, Sendai, Japan
| | - Ritsuko Shimizu
- Tohoku University, Tohoku Medical Megabank Organization, Sendai, Japan
| | - Kei Nakagawa
- Department of Surgery, Tohoku University Graduate School of Medicine, 1-1, Seiryo-Machi, Aoba-Ku, Sendai, 980-8574, Japan
| | - Shinobu Ohnuma
- Department of Surgery, Tohoku University Graduate School of Medicine, 1-1, Seiryo-Machi, Aoba-Ku, Sendai, 980-8574, Japan
| | - Takashi Kamei
- Department of Surgery, Tohoku University Graduate School of Medicine, 1-1, Seiryo-Machi, Aoba-Ku, Sendai, 980-8574, Japan
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, 1-1, Seiryo-Machi, Aoba-Ku, Sendai, 980-8574, Japan
| |
Collapse
|
10
|
Xiang T, Deng Z, Yang C, Tan J, Dou C, Luo F, Chen Y. Bile acid metabolism regulatory network orchestrates bone homeostasis. Pharmacol Res 2023; 196:106943. [PMID: 37777075 DOI: 10.1016/j.phrs.2023.106943] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/18/2023] [Accepted: 09/28/2023] [Indexed: 10/02/2023]
Abstract
Bile acids (BAs), synthesized in the liver and modified by the gut microbiota, have been widely appreciated not only as simple lipid emulsifiers, but also as complex metabolic regulators and momentous signaling molecules, which play prominent roles in the complex interaction among several metabolic systems. Recent studies have drawn us eyes on the diverse physiological functions of BAs, to enlarge the knowledge about the "gut-bone" axis due to the participation about the gut microbiota-derived BAs to modulate bone homeostasis at physiological and pathological stations. In this review, we have summarized the metabolic processes of BAs and highlighted the crucial roles of BAs targeting bile acid-activated receptors, promoting the proliferation and differentiation of osteoblasts (OBs), inhibiting the activity of osteoclasts (OCs), as well as reducing articular cartilage degradation, thus facilitating bone repair. In addition, we have also focused on the bidirectional effects of BA signaling networks in coordinating the dynamic balance of bone matrix and demonstrated the promising effects of BAs on the development or treatment for pathological bone diseases. In a word, further clinical applications targeting BA metabolism or modulating gut metabolome and related derivatives may be developed as effective therapeutic strategies for bone destruction diseases.
Collapse
Affiliation(s)
- Tingwen Xiang
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; College of Basic Medical Science, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Zihan Deng
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Chuan Yang
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jiulin Tan
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Ce Dou
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| | - Fei Luo
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| | - Yueqi Chen
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| |
Collapse
|
11
|
Carson MD, Warner AJ, Geiser VL, Hathaway-Schrader JD, Alekseyenko AV, Marshall J, Westwater C, Novince CM. Prolonged Antibiotic Exposure during Adolescence Dysregulates Liver Metabolism and Promotes Adiposity in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:796-812. [PMID: 36906264 PMCID: PMC10284030 DOI: 10.1016/j.ajpath.2023.02.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/24/2023] [Accepted: 02/16/2023] [Indexed: 03/12/2023]
Abstract
Antibiotic administration during early life has been shown to have lasting effects on the gut microbiota, which have been linked to sustained alterations in liver metabolism and adiposity. Recent investigations have discerned that the gut microbiota continues to develop toward an adult-like profile during adolescence. However, the impact of antibiotic exposure during adolescence on metabolism and adiposity is unclear. Herein, a retrospective analysis of Medicaid claims data was performed, which indicated that tetracycline class antibiotics are commonly prescribed for the systemic treatment of adolescent acne. The purpose of this was to discern the impact of a prolonged tetracycline antibiotic exposure during adolescence on the gut microbiota, liver metabolism, and adiposity. Male C57BL/6T specific pathogen-free mice were administered a tetracycline antibiotic during the pubertal/postpubertal adolescent growth phase. Groups were euthanized at different time points to assess immediate and sustained antibiotic treatment effects. Antibiotic exposure during adolescence caused lasting genera-level shifts in the intestinal bacteriome and persistent dysregulation of metabolic pathways in the liver. Dysregulated hepatic metabolism was linked to sustained disruption of the intestinal farnesoid X receptor-fibroblast growth factor 15 axis, a gut-liver endocrine axis that supports metabolic homeostasis. Antibiotic exposure during adolescence increased subcutaneous, visceral, and marrow adiposity, which intriguingly manifested following antibiotic therapy. This preclinical work highlights that prolonged antibiotic courses for the clinical treatment of adolescent acne may have unintended deleterious effects on liver metabolism and adiposity.
Collapse
Affiliation(s)
- Matthew D Carson
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina; Division of Endocrinology, Department of Pediatrics, College of Medicine, Medical University of South Carolina, Charleston, South Carolina; Division of Periodontics, Department of Stomatology, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Amy J Warner
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina; Division of Endocrinology, Department of Pediatrics, College of Medicine, Medical University of South Carolina, Charleston, South Carolina; Division of Periodontics, Department of Stomatology, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Vincenza L Geiser
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina; Division of Endocrinology, Department of Pediatrics, College of Medicine, Medical University of South Carolina, Charleston, South Carolina; Division of Periodontics, Department of Stomatology, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Jessica D Hathaway-Schrader
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina; Division of Endocrinology, Department of Pediatrics, College of Medicine, Medical University of South Carolina, Charleston, South Carolina; Division of Periodontics, Department of Stomatology, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Alexander V Alekseyenko
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina; Division of Biomedical Informatics Center, Program for Human Microbiome Research, Department of Public Health Sciences, College of Medicine, Medical University of South Carolina, Charleston, South Carolina; Department of Healthcare Leadership and Management, College of Health Professions, Medical University of South Carolina, Charleston, South Carolina
| | - Julie Marshall
- Division of Population Oral Health, Department of Stomatology, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina; Department of Public Health Sciences, College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Caroline Westwater
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina; Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Chad M Novince
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina; Division of Endocrinology, Department of Pediatrics, College of Medicine, Medical University of South Carolina, Charleston, South Carolina; Division of Periodontics, Department of Stomatology, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina.
| |
Collapse
|
12
|
Gillard J, Leclercq IA. Biological tuners to reshape the bile acid pool for therapeutic purposes in non-alcoholic fatty liver disease. Clin Sci (Lond) 2023; 137:65-85. [PMID: 36601783 PMCID: PMC9816373 DOI: 10.1042/cs20220697] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/08/2022] [Accepted: 12/16/2022] [Indexed: 01/06/2023]
Abstract
Bile acids synthesized within the hepatocytes are transformed by gut microorganisms and reabsorbed into the portal circulation. During their enterohepatic cycling, bile acids act as signaling molecules by interacting with receptors to regulate pathways involved in many physiological processes. The bile acid pool, composed of a variety of bile acid species, has been shown to be altered in diseases, hence contributing to disease pathogenesis. Thus, understanding the changes in bile acid pool size and composition in pathological processes will help to elaborate effective pharmacological treatments. Five crucial steps along the enterohepatic cycle shape the bile acid pool size and composition, offering five possible targets for therapeutic intervention. In this review, we provide an insight on the strategies to modulate the bile acid pool, and then we discuss the potential benefits in non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Justine Gillard
- Laboratory of Hepato‐Gastroenterology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
| | - Isabelle A. Leclercq
- Laboratory of Hepato‐Gastroenterology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
13
|
Carson MD, Warner AJ, Hathaway-Schrader JD, Geiser VL, Kim J, Gerasco JE, Hill WD, Lemasters JJ, Alekseyenko AV, Wu Y, Yao H, Aguirre JI, Westwater C, Novince CM. Minocycline-induced disruption of the intestinal FXR/FGF15 axis impairs osteogenesis in mice. JCI Insight 2023; 8:160578. [PMID: 36413391 PMCID: PMC9870091 DOI: 10.1172/jci.insight.160578] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 11/16/2022] [Indexed: 11/24/2022] Open
Abstract
Antibiotic-induced shifts in the indigenous gut microbiota influence normal skeletal maturation. Current theory implies that gut microbiota actions on bone occur through a direct gut/bone signaling axis. However, our prior work supports that a gut/liver signaling axis contributes to gut microbiota effects on bone. Our purpose was to investigate the effects of minocycline, a systemic antibiotic treatment for adolescent acne, on pubertal/postpubertal skeletal maturation. Sex-matched specific pathogen-free (SPF) and germ-free (GF) C57BL/6T mice were administered a clinically relevant minocycline dose from age 6-12 weeks. Minocycline caused dysbiotic shifts in the gut bacteriome and impaired skeletal maturation in SPF mice but did not alter the skeletal phenotype in GF mice. Minocycline administration in SPF mice disrupted the intestinal farnesoid X receptor/fibroblast growth factor 15 axis, a gut/liver endocrine axis supporting systemic bile acid homeostasis. Minocycline-treated SPF mice had increased serum conjugated bile acids that were farnesoid X receptor (FXR) antagonists, suppressed osteoblast function, decreased bone mass, and impaired bone microarchitecture and fracture resistance. Stimulating osteoblasts with the serum bile acid profile from minocycline-treated SPF mice recapitulated the suppressed osteogenic phenotype found in vivo, which was mediated through attenuated FXR signaling. This work introduces bile acids as a potentially novel mediator of gut/liver signaling actions contributing to gut microbiota effects on bone.
Collapse
Affiliation(s)
- Matthew D Carson
- Department of Oral Health Sciences, College of Dental Medicine.,Department of Pediatrics, Division of Endocrinology, College of Medicine.,Department of Stomatology, Division of Periodontics, College of Dental Medicine
| | - Amy J Warner
- Department of Oral Health Sciences, College of Dental Medicine.,Department of Pediatrics, Division of Endocrinology, College of Medicine.,Department of Stomatology, Division of Periodontics, College of Dental Medicine
| | - Jessica D Hathaway-Schrader
- Department of Oral Health Sciences, College of Dental Medicine.,Department of Pediatrics, Division of Endocrinology, College of Medicine.,Department of Stomatology, Division of Periodontics, College of Dental Medicine
| | - Vincenza L Geiser
- Department of Oral Health Sciences, College of Dental Medicine.,Department of Pediatrics, Division of Endocrinology, College of Medicine.,Department of Stomatology, Division of Periodontics, College of Dental Medicine
| | - Joseph Kim
- Department of Oral Health Sciences, College of Dental Medicine.,Department of Pediatrics, Division of Endocrinology, College of Medicine.,Department of Stomatology, Division of Periodontics, College of Dental Medicine
| | - Joy E Gerasco
- Department of Oral Health Sciences, College of Dental Medicine.,Department of Drug Discovery & Biomedical Sciences, College of Pharmacy
| | - William D Hill
- Department of Pathology and Laboratory Medicine, College of Medicine
| | - John J Lemasters
- Department of Drug Discovery & Biomedical Sciences, College of Pharmacy.,Department of Biochemistry & Molecular Biology, College of Medicine
| | - Alexander V Alekseyenko
- Department of Oral Health Sciences, College of Dental Medicine.,Biomedical Informatics Center, Program for Human Microbiome Research, Department of Public Health Sciences, College of Medicine.,Department of Healthcare Leadership and Management, College of Health Professions; and
| | - Yongren Wu
- Department of Orthopedics & Physical Medicine, College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA.,Department of Bioengineering, College of Engineering, Clemson University, Clemson, South Carolina, USA
| | - Hai Yao
- Department of Oral Health Sciences, College of Dental Medicine.,Department of Bioengineering, College of Engineering, Clemson University, Clemson, South Carolina, USA
| | - J Ignacio Aguirre
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
| | - Caroline Westwater
- Department of Oral Health Sciences, College of Dental Medicine.,Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Chad M Novince
- Department of Oral Health Sciences, College of Dental Medicine.,Department of Pediatrics, Division of Endocrinology, College of Medicine.,Department of Stomatology, Division of Periodontics, College of Dental Medicine
| |
Collapse
|
14
|
Sun J, Fan J, Li T, Yan X, Jiang Y. Nuciferine Protects Against High-Fat Diet-Induced Hepatic Steatosis via Modulation of Gut Microbiota and Bile Acid Metabolism in Rats. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:12014-12028. [PMID: 36106619 DOI: 10.1021/acs.jafc.2c04817] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Our previous study showed that nuciferine (NF) attenuated non-alcoholic fatty liver disease (NAFLD), which is attributed to a high-fat diet (HFD) through reinforcing intestinal barrier functions, regulating lipid metabolism, and improving inflammation. To clarify whether other mechanisms contribute to the anti-NAFLD efficacy of NF, the present study investigated the influence of NF on bile acid (BA) metabolism and gut microbiota in HFD-fed rats. The data demonstrated that NF changed the composition of colonic BA, particularly elevating conjugated BA and non-12OH BA levels. As shown by downregulated protein levels of FXR, FGF15, FGFR4, and ASBT and upregulated protein levels of CYP7A1 and CYP27A1, NF inhibited ileal FXR signaling, promoted BA synthesis, suppressed BA reabsorption, and facilitated fecal BA excretion. NF might affect hepatic FXR signaling, BA conjugation, and enterohepatic circulation by the changed mRNA levels of Fxr, Shp, Baat, Bacs, Bsep, Ntcp, Ibabp, and Ostα/β. Meanwhile, NF regulated the gut microbiota, characterized by decreased BSH-producing genus, 7α-dehydroxylation genus, and increased taurine metabolism-related genus. Spearman rank correlation analysis implied that Colidextribacter, Adlercreutzia, Family_XIII_AD3011_group, Lachnospiraceae_UCG-010, Eisenbergiella, and UCG-005 were robustly associated with particular BA monomers. In conclusion, our experiment results suggested that NF could exert a mitigating effect on NAFLD via regulating BA metabolism and modulating the gut microbiota.
Collapse
Affiliation(s)
- Jingyue Sun
- College of Biosystems Engineering and Food Science, Zhejiang University, No.866 Yuhangtang Road, Xihu District, Hangzhou 310058, China
| | - Jiemin Fan
- College of Biosystems Engineering and Food Science, Zhejiang University, No.866 Yuhangtang Road, Xihu District, Hangzhou 310058, China
| | - Tingting Li
- College of Biosystems Engineering and Food Science, Zhejiang University, No.866 Yuhangtang Road, Xihu District, Hangzhou 310058, China
| | - Xiaoxue Yan
- College of Biosystems Engineering and Food Science, Zhejiang University, No.866 Yuhangtang Road, Xihu District, Hangzhou 310058, China
| | - Yihong Jiang
- College of Biosystems Engineering and Food Science, Zhejiang University, No.866 Yuhangtang Road, Xihu District, Hangzhou 310058, China
| |
Collapse
|
15
|
Molecular Basis of Bile Acid-FXR-FGF15/19 Signaling Axis. Int J Mol Sci 2022; 23:ijms23116046. [PMID: 35682726 PMCID: PMC9181207 DOI: 10.3390/ijms23116046] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 02/04/2023] Open
Abstract
Bile acids (BAs) are a group of amphiphilic molecules consisting of a rigid steroid core attached to a hydroxyl group with a varying number, position, and orientation, and a hydrophilic side chain. While BAs act as detergents to solubilize lipophilic nutrients in the small intestine during digestion and absorption, they also act as hormones. Farnesoid X receptor (FXR) is a nuclear receptor that forms a heterodimer with retinoid X receptor α (RXRα), is activated by BAs in the enterohepatic circulation reabsorbed via transporters in the ileum and the colon, and plays a critical role in regulating gene expression involved in cholesterol, BA, and lipid metabolism in the liver. The FXR/RXRα heterodimer also exists in the distal ileum and regulates production of fibroblast growth factor (FGF) 15/FGF19, a hormone traveling via the enterohepatic circulation that activates hepatic FGF receptor 4 (FGFR4)-β-klotho receptor complex and regulates gene expression involved in cholesterol, BA, and lipid metabolism, as well as those regulating cell proliferation. Agonists for FXR and analogs for FGF15/19 are currently recognized as a promising therapeutic target for metabolic syndrome and cholestatic diseases.
Collapse
|
16
|
Li Y, Hou H, Wang X, Dai X, Zhang W, Tang Q, Dong Y, Yan C, Wang B, Li Z, Cao H. Diammonium Glycyrrhizinate Ameliorates Obesity Through Modulation of Gut Microbiota-Conjugated BAs-FXR Signaling. Front Pharmacol 2022; 12:796590. [PMID: 34992541 PMCID: PMC8724542 DOI: 10.3389/fphar.2021.796590] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity is a worldwide epidemic metabolic disease. Gut microbiota dysbiosis and bile acids (BAs) metabolism disorder are closely related to obesity. Farnesoid X-activated receptor (FXR), served as a link between gut microbiota and BAs, is involved in maintaining metabolic homeostasis and regulating glucose and lipid metabolism. We previously reported that diammonium glycyrrhizinate (DG) could alter gut microbiota and prevent non-alcoholic fatty liver disease. However, it remains ambiguous how DG affects the gut microbiota to regulate host metabolism. In this present study, 16S rRNA Illumina NovaSeq and metabolomic analysis revealed that DG treatment suppressed microbes associated with bile-salt hydrolase (BSH) activity, which, in turn, increased the levels of taurine-conjugated BAs accompanied by inhibition of ileal FXR-FGF15 signaling. As a result, several obesity-related metabolism were improved, like lower serum glucose and insulin levels, increased insulin sensitivity, few hepatic steatosis and resistance to weight gain. Additionally, decreased level of serum lipopolysaccharide was observed, which contributed to a strengthened intestinal barrier. The effect of DG on weight loss was slightly enhanced in the antibiotics-treated obese mice. Collectively, the efficacy of DG in the treatment of obesity might depend on gut microbiota-conjugated BAs-FXR axis. Hence, it will provide a potential novel approach for the treatment of obesity.
Collapse
Affiliation(s)
- Yun Li
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China.,Department of Pharmacy, General Hospital, Tianjin Medical University, Tianjin, China
| | - Huiqin Hou
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Xianglu Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Xin Dai
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Wanru Zhang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Qiang Tang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Yue Dong
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Chen Yan
- Department of Pharmacy, General Hospital, Tianjin Medical University, Tianjin, China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Zhengxiang Li
- Department of Pharmacy, General Hospital, Tianjin Medical University, Tianjin, China
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
17
|
Miyata M, Tanaka T, Takahashi K, Funaki A, Sugiura Y. Cholesterol-lowering effects of taurine through the reduction of ileal FXR signaling due to the alteration of ileal bile acid composition. Amino Acids 2021; 53:1523-1532. [PMID: 34596761 DOI: 10.1007/s00726-021-03068-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 08/14/2021] [Indexed: 01/05/2023]
Abstract
Studies using animal models of hypercholesterolemia have established that taurine reduces cholesterol levels; however, the precise mechanism underlying this cholesterol-lowering effect is unclear. This study addressed this issue by investigating whether bile acid/farnesoid X receptor (FXR) signaling is involved in taurine-mediated cholesterol-lowering effect. Fxr-null and wild-type mice were administered 2% (w/v) taurine in their drinking water and fed a control diet or control diet supplemented with 1% (w/w) cholesterol (cholesterol diet) for 10 days. Taurine intake did not significantly alter hepatic and serum total cholesterol (TC) levels and bile acid compositions of the liver and intestinal lumen in Fxr-null and wild-type mice fed the control diet. By changing to a cholesterol diet, taurine intake significantly decreased hepatic and serum cholesterol levels in wild-type mice. In contrast, it significantly decreased hepatic, not serum, cholesterol levels in Fxr-null mice. Taurine intake significantly altered the bile acid composition of the intestinal lumen in wild-type mice fed a cholesterol diet, but not in Fxr-null mice. An increase in FXR antagonistic bile acids was detected in the intestinal lumen of taurine-treated wild-type mice fed a cholesterol diet. Taurine intake reduced the ileal expression of FXR target genes fibroblast growth factor 15 (Fgf15) and small heterodimer partner (Shp). In contrast, it enhanced the hepatic expression of cholesterol 7α-hydroxylase (Cyp7a1) in wild-type mice fed a cholesterol diet, but not in Fxr-null mice. These results suggest that taurine is partially involved in cholesterol lowering by reducing the ileal FXR signaling due to the alteration of ileal bile acid composition.
Collapse
Affiliation(s)
- Masaaki Miyata
- Department of Food Science and Technology, National Research and Development Agency, Japan Fisheries Research and Education Agency, National Fisheries University, 2-7-1, Nagata-honmachi, Shimonoseki, 759-6595, Japan.
| | - Tomoyuki Tanaka
- Department of Food Science and Technology, National Research and Development Agency, Japan Fisheries Research and Education Agency, National Fisheries University, 2-7-1, Nagata-honmachi, Shimonoseki, 759-6595, Japan
| | - Kazuho Takahashi
- Department of Food Science and Technology, National Research and Development Agency, Japan Fisheries Research and Education Agency, National Fisheries University, 2-7-1, Nagata-honmachi, Shimonoseki, 759-6595, Japan
| | - Akihiro Funaki
- Department of Food Science and Technology, National Research and Development Agency, Japan Fisheries Research and Education Agency, National Fisheries University, 2-7-1, Nagata-honmachi, Shimonoseki, 759-6595, Japan
| | - Yoshimasa Sugiura
- Department of Food Science and Technology, National Research and Development Agency, Japan Fisheries Research and Education Agency, National Fisheries University, 2-7-1, Nagata-honmachi, Shimonoseki, 759-6595, Japan
| |
Collapse
|
18
|
Xu M, Cen M, Shen Y, Zhu Y, Cheng F, Tang L, Hu W, Dai N. Deoxycholic Acid-Induced Gut Dysbiosis Disrupts Bile Acid Enterohepatic Circulation and Promotes Intestinal Inflammation. Dig Dis Sci 2021; 66:568-576. [PMID: 32198567 DOI: 10.1007/s10620-020-06208-3] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 03/11/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND A Western diet is a risk factor for the development of inflammatory bowel disease (IBD). High levels of fecal deoxycholic acid (DCA) in response to a Western diet contribute to bowel inflammatory injury. However, the mechanism of DCA in the natural course of IBD development remains unanswered. AIMS The aim of this study is to investigate the effect of DCA on the induction of gut dysbiosis and its roles in the development of intestinal inflammation. METHODS Wild-type C57BL/6J mice were fed an AIN-93G diet, either supplemented with or without 0.2% DCA, and killed at 24 weeks. Distal ileum and colon tissues were assessed by histopathological analysis. Hepatic and ileal gene expression was examined by qPCR, and the gut microbiota was analyzed by high-throughput 16S rRNA gene sequencing. HPLC-MS was used for fecal bile acid quantification. RESULTS Mice fed the DCA-supplemented diet developed focal areas of ileal and colonic inflammation, accompanied by alteration of the composition of the intestinal microbiota and accumulation of fecal bile acids. DCA-induced dysbiosis decreased the deconjugation of bile acids, and this regulation was associated with the repressed expression of target genes in the enterohepatic farnesoid X receptor-fibroblast growth factor (FXR-FGF15) axis, leading to upregulation of hepatic de novo bile acid synthesis. CONCLUSIONS These results suggest that DCA-induced gut dysbiosis may act as a key etiologic factor in intestinal inflammation, associated with bile acid metabolic disturbance and downregulation of the FXR-FGF15 axis.
Collapse
Affiliation(s)
- Mengque Xu
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
| | - Mengsha Cen
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
| | - Yuqin Shen
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
| | - Yubin Zhu
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
| | - Fangli Cheng
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
| | - Linlin Tang
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
| | - Weiling Hu
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China
| | - Ning Dai
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, No. 3 Qingchun East Road, Jianggan District, Hangzhou, 310016, China.
| |
Collapse
|
19
|
Engevik MA, Danhof HA, Chang-Graham AL, Spinler JK, Engevik KA, Herrmann B, Endres BT, Garey KW, Hyser JM, Britton RA, Versalovic J. Human intestinal enteroids as a model of Clostridioides difficile-induced enteritis. Am J Physiol Gastrointest Liver Physiol 2020; 318:G870-G888. [PMID: 32223302 PMCID: PMC7272722 DOI: 10.1152/ajpgi.00045.2020] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Clostridioides difficile is an important nosocomial pathogen that produces toxins to cause life-threatening diarrhea and colitis. Toxins bind to epithelial receptors and promote the collapse of the actin cytoskeleton. C. difficile toxin activity is commonly studied in cancer-derived and immortalized cell lines. However, the biological relevance of these models is limited. Moreover, no model is available for examining C. difficile-induced enteritis, an understudied health problem. We hypothesized that human intestinal enteroids (HIEs) express toxin receptors and provide a new model to dissect C. difficile cytotoxicity in the small intestine. We generated biopsy-derived jejunal HIE and Vero cells, which stably express LifeAct-Ruby, a fluorescent label of F-actin, to monitor actin cytoskeleton rearrangement by live-cell microscopy. Imaging analysis revealed that toxins from pathogenic C. difficile strains elicited cell rounding in a strain-dependent manner, and HIEs were tenfold more sensitive to toxin A (TcdA) than toxin B (TcdB). By quantitative PCR, we paradoxically found that HIEs expressed greater quantities of toxin receptor mRNA and yet exhibited decreased sensitivity to toxins when compared with traditionally used cell lines. We reasoned that these differences may be explained by components, such as mucins, that are present in HIEs cultures, that are absent in immortalized cell lines. Addition of human-derived mucin 2 (MUC2) to Vero cells delayed cell rounding, indicating that mucus serves as a barrier to toxin-receptor binding. This work highlights that investigation of C. difficile infection in that HIEs can provide important insights into the intricate interactions between toxins and the human intestinal epithelium.NEW & NOTEWORTHY In this article, we developed a novel model of Clostridioides difficile-induced enteritis using jejunal-derived human intestinal enteroids (HIEs) transduced with fluorescently tagged F-actin. Using live-imaging, we identified that jejunal HIEs express high levels of TcdA and CDT receptors, are more sensitive to TcdA than TcdB, and secrete mucus, which delays toxin-epithelial interactions. This work also optimizes optically clear C. difficile-conditioned media suitable for live-cell imaging.
Collapse
Affiliation(s)
- Melinda A. Engevik
- 1Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas,2Department of Pathology, Texas Children’s Hospital, Houston, Texas
| | - Heather A. Danhof
- 3Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, Houston, Texas,4Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | | | - Jennifer K. Spinler
- 1Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas,2Department of Pathology, Texas Children’s Hospital, Houston, Texas
| | - Kristen A. Engevik
- 3Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, Houston, Texas,4Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | - Beatrice Herrmann
- 1Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas,2Department of Pathology, Texas Children’s Hospital, Houston, Texas
| | - Bradley T. Endres
- 5Department of Pharmacy Practice and Translational Research, University of Houston College of Pharmacy, Houston, Texas
| | - Kevin W. Garey
- 5Department of Pharmacy Practice and Translational Research, University of Houston College of Pharmacy, Houston, Texas
| | - Joseph M. Hyser
- 1Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas,2Department of Pathology, Texas Children’s Hospital, Houston, Texas
| | - Robert A. Britton
- 3Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, Houston, Texas,4Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | - James Versalovic
- 1Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas,2Department of Pathology, Texas Children’s Hospital, Houston, Texas
| |
Collapse
|
20
|
Bogatyrev SR, Rolando JC, Ismagilov RF. Self-reinoculation with fecal flora changes microbiota density and composition leading to an altered bile-acid profile in the mouse small intestine. MICROBIOME 2020; 8:19. [PMID: 32051033 PMCID: PMC7017497 DOI: 10.1186/s40168-020-0785-4] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/05/2020] [Indexed: 05/06/2023]
Abstract
BACKGROUND The upper gastrointestinal tract plays a prominent role in human physiology as the primary site for enzymatic digestion and nutrient absorption, immune sampling, and drug uptake. Alterations to the small intestine microbiome have been implicated in various human diseases, such as non-alcoholic steatohepatitis and inflammatory bowel conditions. Yet, the physiological and functional roles of the small intestine microbiota in humans remain poorly characterized because of the complexities associated with its sampling. Rodent models are used extensively in microbiome research and enable the spatial, temporal, compositional, and functional interrogation of the gastrointestinal microbiota and its effects on the host physiology and disease phenotype. Classical, culture-based studies have documented that fecal microbial self-reinoculation (via coprophagy) affects the composition and abundance of microbes in the murine proximal gastrointestinal tract. This pervasive self-reinoculation behavior could be a particularly relevant study factor when investigating small intestine microbiota. Modern microbiome studies either do not take self-reinoculation into account, or assume that approaches such as single housing mice or housing on wire mesh floors eliminate it. These assumptions have not been rigorously tested with modern tools. Here, we used quantitative 16S rRNA gene amplicon sequencing, quantitative microbial functional gene content inference, and metabolomic analyses of bile acids to evaluate the effects of self-reinoculation on microbial loads, composition, and function in the murine upper gastrointestinal tract. RESULTS In coprophagic mice, continuous self-exposure to the fecal flora had substantial quantitative and qualitative effects on the upper gastrointestinal microbiome. These differences in microbial abundance and community composition were associated with an altered profile of the small intestine bile acid pool, and, importantly, could not be inferred from analyzing large intestine or stool samples. Overall, the patterns observed in the small intestine of non-coprophagic mice (reduced total microbial load, low abundance of anaerobic microbiota, and bile acids predominantly in the conjugated form) resemble those typically seen in the human small intestine. CONCLUSIONS Future studies need to take self-reinoculation into account when using mouse models to evaluate gastrointestinal microbial colonization and function in relation to xenobiotic transformation and pharmacokinetics or in the context of physiological states and diseases linked to small intestine microbiome and to small intestine dysbiosis. Video abstract.
Collapse
Affiliation(s)
- Said R Bogatyrev
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Justin C Rolando
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 1200 E. California Blvd, Pasadena, CA, USA
| | - Rustem F Ismagilov
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 1200 E. California Blvd, Pasadena, CA, USA.
| |
Collapse
|
21
|
Zhang Y, Bobe G, Revel JS, Rodrigues R, Sharpton TJ, Fantacone ML, Raslan K, Miranda CL, Lowry MB, Blakemore PR, Morgun A, Shulzhenko N, Maier CS, Stevens JF, Gombart AF. Improvements in Metabolic Syndrome by Xanthohumol Derivatives Are Linked to Altered Gut Microbiota and Bile Acid Metabolism. Mol Nutr Food Res 2020; 64:e1900789. [PMID: 31755244 PMCID: PMC7029812 DOI: 10.1002/mnfr.201900789] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/21/2019] [Indexed: 12/21/2022]
Abstract
SCOPE Two hydrogenated xanthohumol (XN) derivatives, α,β-dihydro-XN (DXN) and tetrahydro-XN (TXN), improved parameters of metabolic syndrome (MetS), a critical risk factor of cardiovascular disease (CVD) and type 2 diabetes, in a diet-induced obese murine model. It is hypothesized that improvements in obesity and MetS are linked to changes in composition of the gut microbiota, bile acid metabolism, intestinal barrier function, and inflammation. METHODS AND RESULTS To test this hypothesis, 16S rRNA genes were sequenced and bile acids were measured in fecal samples from C57BL/6J mice fed a high-fat diet (HFD) or HFD containing XN, DXN or TXN. Expression of genes associated with epithelial barrier function, inflammation, and bile acid metabolism were measured in the colon, white adipose tissue (WAT), and liver, respectively. Administration of XN derivatives decreases intestinal microbiota diversity and abundance-specifically Bacteroidetes and Tenericutes-alters bile acid metabolism, and reduces inflammation. In WAT, TXN supplementation decreases pro-inflammatory gene expression by suppressing macrophage infiltration. Transkingdom network analysis connects changes in the microbiota to improvements in MetS in the host. CONCLUSION Changes in the gut microbiota and bile acid metabolism may explain, in part, the improvements in obesity and MetS associated with administration of XN and its derivatives.
Collapse
Affiliation(s)
- Yang Zhang
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon, 97331, USA
- School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, 97331, USA
| | - Gerd Bobe
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon, 97331, USA
- Department of Animal Sciences, Oregon State University, Corvallis, Oregon, 97331, USA
| | - Johana S. Revel
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon, 97331, USA
- Department of Chemistry, Oregon State University, Corvallis, Oregon, 97331, USA
| | - Richard Rodrigues
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, Oregon, 97331, USA
| | - Thomas J. Sharpton
- Department of Microbiology, Oregon State University, Corvallis, Oregon, 97331, USA
- Department of Statistics, Oregon State University, Corvallis, Oregon, 97331, USA
| | - Mary L. Fantacone
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon, 97331, USA
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon, 97331, USA
| | - Kareem Raslan
- Department of Microbiology, Oregon State University, Corvallis, Oregon, 97331, USA
| | - Cristobal L. Miranda
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon, 97331, USA
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, Oregon, 97331, USA
| | - Malcolm B. Lowry
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon, 97331, USA
- Department of Microbiology, Oregon State University, Corvallis, Oregon, 97331, USA
| | - Paul R. Blakemore
- Department of Chemistry, Oregon State University, Corvallis, Oregon, 97331, USA
| | - Andrey Morgun
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, Oregon, 97331, USA
| | - Natalia Shulzhenko
- College of Veterinary Medicine; Oregon State University, Corvallis, Oregon, 97331, USA
| | - Claudia S. Maier
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon, 97331, USA
- Department of Chemistry, Oregon State University, Corvallis, Oregon, 97331, USA
| | - Jan F. Stevens
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon, 97331, USA
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, Oregon, 97331, USA
| | - Adrian F. Gombart
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon, 97331, USA
- School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, 97331, USA
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon, 97331, USA
| |
Collapse
|
22
|
Yu Y, Raka F, Adeli K. The Role of the Gut Microbiota in Lipid and Lipoprotein Metabolism. J Clin Med 2019; 8:jcm8122227. [PMID: 31861086 PMCID: PMC6947520 DOI: 10.3390/jcm8122227] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/06/2019] [Accepted: 12/08/2019] [Indexed: 12/12/2022] Open
Abstract
Both environmental and genetic factors contribute to relative species abundance and metabolic characteristics of the intestinal microbiota. The intestinal microbiota and accompanying microbial metabolites differ substantially in those who are obese or have other metabolic disorders. Accumulating evidence from germ-free mice and antibiotic-treated animal models suggests that altered intestinal gut microbiota contributes significantly to metabolic disorders involving impaired glucose and lipid metabolism. This review will summarize recent findings on potential mechanisms by which the microbiota affects intestinal lipid and lipoprotein metabolism including microbiota dependent changes in bile acid metabolism which affects bile acid signaling by bile acid receptors FXR and TGR5. Microbiota changes also involve altered short chain fatty acid signaling and influence enteroendocrine cell function including GLP-1/GLP-2-producing L-cells which regulate postprandial lipid metabolism.
Collapse
Affiliation(s)
- Yijing Yu
- Molecular Medicine, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; (Y.Y.); (F.R.)
| | - Fitore Raka
- Molecular Medicine, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; (Y.Y.); (F.R.)
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Khosrow Adeli
- Molecular Medicine, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; (Y.Y.); (F.R.)
- Departments of Laboratory Medicine & Pathobiology and Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
- Correspondence: ; Tel.: +416-813-8682
| |
Collapse
|
23
|
Huang F, Zheng X, Ma X, Jiang R, Zhou W, Zhou S, Zhang Y, Lei S, Wang S, Kuang J, Han X, Wei M, You Y, Li M, Li Y, Liang D, Liu J, Chen T, Yan C, Wei R, Rajani C, Shen C, Xie G, Bian Z, Li H, Zhao A, Jia W. Theabrownin from Pu-erh tea attenuates hypercholesterolemia via modulation of gut microbiota and bile acid metabolism. Nat Commun 2019; 10:4971. [PMID: 31672964 PMCID: PMC6823360 DOI: 10.1038/s41467-019-12896-x] [Citation(s) in RCA: 502] [Impact Index Per Article: 83.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 10/03/2019] [Indexed: 12/16/2022] Open
Abstract
Pu-erh tea displays cholesterol-lowering properties, but the underlying mechanism has not been elucidated. Theabrownin is one of the most active and abundant pigments in Pu-erh tea. Here, we show that theabrownin alters the gut microbiota in mice and humans, predominantly suppressing microbes associated with bile-salt hydrolase (BSH) activity. Theabrownin increases the levels of ileal conjugated bile acids (BAs) which, in turn, inhibit the intestinal FXR-FGF15 signaling pathway, resulting in increased hepatic production and fecal excretion of BAs, reduced hepatic cholesterol, and decreased lipogenesis. The inhibition of intestinal FXR-FGF15 signaling is accompanied by increased gene expression of enzymes in the alternative BA synthetic pathway, production of hepatic chenodeoxycholic acid, activation of hepatic FXR, and hepatic lipolysis. Our results shed light into the mechanisms behind the cholesterol- and lipid-lowering effects of Pu-erh tea, and suggest that decreased intestinal BSH microbes and/or decreased FXR-FGF15 signaling may be potential anti-hypercholesterolemia and anti-hyperlipidemia therapies.
Collapse
Affiliation(s)
- Fengjie Huang
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaojiao Zheng
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xiaohui Ma
- Department of Pharmacology and Toxicology, Tasly Pharmaceutical Co. Ltd, Tianjin, China
| | - Runqiu Jiang
- University of Hawaii Cancer Center, Honolulu, USA
| | - Wangyi Zhou
- Department of Pharmacology and Toxicology, Tasly Pharmaceutical Co. Ltd, Tianjin, China
| | - Shuiping Zhou
- Department of Pharmacology and Toxicology, Tasly Pharmaceutical Co. Ltd, Tianjin, China
| | - Yunjing Zhang
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Sha Lei
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Shouli Wang
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Junliang Kuang
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xiaolong Han
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Meilin Wei
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yijun You
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Mengci Li
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yitao Li
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Dandan Liang
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jiajian Liu
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Tianlu Chen
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Chao Yan
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Runmin Wei
- University of Hawaii Cancer Center, Honolulu, USA
| | | | - Chengxing Shen
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Six People's Hospital, Shanghai, China
| | - Guoxiang Xie
- University of Hawaii Cancer Center, Honolulu, USA
| | - Zhaoxiang Bian
- Chinese Medicine Clinical Study Center, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Houkai Li
- Functional Metabolomic and Gut Microbiome Laboratory, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Aihua Zhao
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| | - Wei Jia
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
- University of Hawaii Cancer Center, Honolulu, USA.
- Chinese Medicine Clinical Study Center, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
| |
Collapse
|
24
|
Debédat J, Amouyal C, Aron-Wisnewsky J, Clément K. Impact of bariatric surgery on type 2 diabetes: contribution of inflammation and gut microbiome? Semin Immunopathol 2019; 41:461-475. [PMID: 31025085 DOI: 10.1007/s00281-019-00738-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 03/15/2019] [Indexed: 02/06/2023]
|
25
|
Intestinal Bile Acids Induce a Morphotype Switch in Vancomycin-Resistant Enterococcus that Facilitates Intestinal Colonization. Cell Host Microbe 2019; 25:695-705.e5. [PMID: 31031170 DOI: 10.1016/j.chom.2019.03.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 01/25/2019] [Accepted: 03/18/2019] [Indexed: 12/22/2022]
Abstract
Vancomycin-resistant Enterococcus (VRE) are highly antibiotic-resistant and readily transmissible pathogens that cause severe infections in hospitalized patients. We discovered that lithocholic acid (LCA), a secondary bile acid prevalent in the cecum and colon of mice and humans, impairs separation of growing VRE diplococci, causing the formation of long chains and increased biofilm formation. Divalent cations reversed this LCA-induced switch to chaining and biofilm formation. Experimental evolution in the presence of LCA yielded mutations in the essential two-component kinase yycG/walK and three-component response regulator liaR that locked VRE in diplococcal mode, impaired biofilm formation, and increased susceptibility to the antibiotic daptomycin. These mutant VRE strains were deficient in host colonization because of their inability to compete with intestinal microbiota. This morphotype switch presents a potential non-bactericidal therapeutic target that may help clear VRE from the intestines of dominated patients, as occurs frequently during hematopoietic stem cell transplantation.
Collapse
|
26
|
Khan TJ, Hasan MN, Azhar EI, Yasir M. Association of gut dysbiosis with intestinal metabolites in response to antibiotic treatment. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.humic.2018.11.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
27
|
Somm E, Jornayvaz FR. Fibroblast Growth Factor 15/19: From Basic Functions to Therapeutic Perspectives. Endocr Rev 2018; 39:960-989. [PMID: 30124818 DOI: 10.1210/er.2018-00134] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 07/10/2018] [Indexed: 12/11/2022]
Abstract
Discovered 20 years ago, fibroblast growth factor (FGF)19, and its mouse ortholog FGF15, were the first members of a new subfamily of FGFs able to act as hormones. During fetal life, FGF15/19 is involved in organogenesis, affecting the development of the ear, eye, heart, and brain. At adulthood, FGF15/19 is mainly produced by the ileum, acting on the liver to repress hepatic bile acid synthesis and promote postprandial nutrient partitioning. In rodents, pharmacologic doses of FGF19 induce the same antiobesity and antidiabetic actions as FGF21, with these metabolic effects being partly mediated by the brain. However, activation of hepatocyte proliferation by FGF19 has long been a challenge to its therapeutic use. Recently, genetic reengineering of the molecule has resolved this issue. Despite a global overlap in expression pattern and function, murine FGF15 and human FGF19 exhibit several differences in terms of regulation, molecular structure, signaling, and biological properties. As most of the knowledge originates from the use of FGF19 in murine models, differences between mice and humans in the biology of FGF15/19 have to be considered for a successful translation from bench to bedside. This review summarizes the basic knowledge concerning FGF15/19 in mice and humans, with a special focus on regulation of production, morphogenic properties, hepatocyte growth, bile acid homeostasis, as well as actions on glucose, lipid, and energy homeostasis. Moreover, implications and therapeutic perspectives concerning FGF19 in human diseases (including obesity, type 2 diabetes, hepatic steatosis, biliary disorders, and cancer) are also discussed.
Collapse
Affiliation(s)
- Emmanuel Somm
- Service of Endocrinology, Diabetes, Hypertension, and Nutrition, Geneva University Hospitals, University of Geneva Medical School, Geneva, Switzerland
| | - François R Jornayvaz
- Service of Endocrinology, Diabetes, Hypertension, and Nutrition, Geneva University Hospitals, University of Geneva Medical School, Geneva, Switzerland
| |
Collapse
|
28
|
Somm E, Henry H, Bruce SJ, Bonnet N, Montandon SA, Niederländer NJ, Messina A, Aeby S, Rosikiewicz M, Fajas L, Sempoux C, Ferrari SL, Greub G, Pitteloud N. β-Klotho deficiency shifts the gut-liver bile acid axis and induces hepatic alterations in mice. Am J Physiol Endocrinol Metab 2018; 315:E833-E847. [PMID: 29944388 DOI: 10.1152/ajpendo.00182.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
β-Klotho (encoded by Klb) is an obligate coreceptor, mediating both fibroblast growth factor (FGF)15 and FGF21 signaling. Klb-/- mice are refractory to metabolic FGF15 and FGF21 action and exhibit derepressed (increased) bile acid (BA) synthesis. Here, we deeply phenotyped male Klb-/- mice on a pure C57BL/6J genetic background, fed a chow diet focusing on metabolic aspects. This aims to better understand the physiological consequences of concomitant FGF15 and FGF21 signaling deficiency, in particular on the gut-liver axis. Klb-/- mice present permanent growth restriction independent of adiposity and energy balance. Klb-/- mice also exhibit few changes in carbohydrate metabolism, combining normal gluco-tolerance, insulin sensitivity, and fasting response with increased gluconeogenic capacity and decreased glycogen mobilization. Livers of Klb-/- mice reveal pathologic features, including a proinflammatory status and initiation of fibrosis. These defects are associated to a massive shift in BA composition in the enterohepatic system and blood circulation featured by a large excess of microbiota-derived deoxycholic acid, classically known for its genotoxicity in the gastrointestinal tract. In conclusion, β-Klotho is a gatekeeper of hepatic integrity through direct action (mediating FGF21 anti-inflammatory signaling) and indirect mechanisms (mediating FGF15 signaling that maintains BA level and composition).
Collapse
Affiliation(s)
- Emmanuel Somm
- Service of Endocrinology, Diabetology, and Metabolism, Department of Physiology, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne , Lausanne , Switzerland
| | - Hugues Henry
- Clinical Chemistry Laboratory, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne , Lausanne , Switzerland
| | - Stephen J Bruce
- Clinical Chemistry Laboratory, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne , Lausanne , Switzerland
| | - Nicolas Bonnet
- Division of Bone Diseases, Department of Internal Medicine Specialties, Geneva University Hospital and Faculty of Medicine , Geneva , Switzerland
| | - Sophie A Montandon
- Service of Endocrinology, Diabetes, Hypertension, and Nutrition, Geneva University Hospital and Faculty of Medicine , Geneva , Switzerland
| | - Nicolas J Niederländer
- Service of Endocrinology, Diabetology, and Metabolism, Department of Physiology, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne , Lausanne , Switzerland
| | - Andrea Messina
- Service of Endocrinology, Diabetology, and Metabolism, Department of Physiology, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne , Lausanne , Switzerland
| | - Sébastien Aeby
- Institute of Microbiology, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne , Lausanne , Switzerland
| | - Marta Rosikiewicz
- Institute of Microbiology, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne , Lausanne , Switzerland
| | - Lluis Fajas
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne , Lausanne , Switzerland
| | - Christine Sempoux
- Institute of Pathology, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne , Lausanne , Switzerland
| | - Serge L Ferrari
- Division of Bone Diseases, Department of Internal Medicine Specialties, Geneva University Hospital and Faculty of Medicine , Geneva , Switzerland
| | - Gilbert Greub
- Institute of Microbiology, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne , Lausanne , Switzerland
| | - Nelly Pitteloud
- Service of Endocrinology, Diabetology, and Metabolism, Department of Physiology, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne , Lausanne , Switzerland
| |
Collapse
|
29
|
Stipanuk MH, Jurkowska H, Niewiadomski J, Mazor KM, Roman HB, Hirschberger LL. Identification of Taurine-Responsive Genes in Murine Liver Using the Cdo1-Null Mouse Model. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 975 Pt 1:475-495. [PMID: 28849476 DOI: 10.1007/978-94-024-1079-2_38] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The cysteine dioxygenase (Cdo1)-null mouse is unable to synthesize hypotaurine and taurine by the cysteine/cysteine sulfinate pathway and has very low taurine levels in all tissues. The lack of taurine is associated with a lack of taurine conjugation of bile acids, a dramatic increase in the total and unconjugated hepatic bile acid pools, and an increase in betaine and other molecules that serve as organic osmolytes. We used the Cdo1-mouse model to determine the effects of taurine deficiency on expression of proteins involved in sulfur amino acid and bile acid metabolism. We identified cysteine sulfinic acid decarboxylase (Csad), betaine:homocysteine methytransferase (Bhmt), cholesterol 7α-hydroxylase (Cyp7a1), and cytochrome P450 3A11 (Cyp3a11) as genes whose hepatic expression is strongly regulated in response to taurine depletion in the Cdo1-null mouse. Dietary taurine supplementation of Cdo1-null mice restored hepatic levels of these four proteins and their respective mRNAs to wild-type levels, whereas dietary taurine supplementation had no effect on abundance of these proteins or mRNAs in wild-type mice.
Collapse
Affiliation(s)
- Martha H Stipanuk
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA.
| | - Halina Jurkowska
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
- Chair of Medical Biochemistry, Jagiellonian University Medical College, Kraków, Poland
| | | | - Kevin M Mazor
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Heather B Roman
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | | |
Collapse
|
30
|
Underrated enemy - from nonalcoholic fatty liver disease to cancers of the gastrointestinal tract. Clin Exp Hepatol 2018; 4:55-71. [PMID: 29904722 PMCID: PMC6000748 DOI: 10.5114/ceh.2018.75955] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 04/17/2018] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is intrahepatic ectopic lipid deposition which is present despite a lack of other causes of secondary hepatic fat accumulation. It is the most common chronic liver disorder in the welldeveloped countries. NAFLD is a multidisciplinary disease that affects various systems and organs and is inextricably linked to simple obesity, metabolic syndrome, insulin resistance and overt diabetes mellitus type 2. The positive energy balance related to obesity leads to a variety of systemic changes including modified levels of insulin, insulin- like growth factor-1, adipokines, hepatokines and cytokines. It is strongly linked to carcinogenesis and new evidence proves that NAFLD is associated with higher risk of all-cause mortality and cancer-specific mortality among cancer survivors. This article focuses on the association between NAFLD and extrahepatic gastrointestinal tract cancers, aiming to shed light on the pathomechanism of changes leading to the development of tumors.
Collapse
|
31
|
Caputi V, Marsilio I, Filpa V, Cerantola S, Orso G, Bistoletti M, Paccagnella N, De Martin S, Montopoli M, Dall'Acqua S, Crema F, Di Gangi I, Galuppini F, Lante I, Bogialli S, Rugge M, Debetto P, Giaroni C, Giron MC. Antibiotic-induced dysbiosis of the microbiota impairs gut neuromuscular function in juvenile mice. Br J Pharmacol 2017; 174:3623-3639. [PMID: 28755521 PMCID: PMC5610159 DOI: 10.1111/bph.13965] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 07/18/2017] [Accepted: 07/19/2017] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE Gut microbiota is essential for the development of the gastrointestinal system, including the enteric nervous system (ENS). Perturbations of gut microbiota in early life have the potential to alter neurodevelopment leading to functional bowel disorders later in life. We examined the hypothesis that gut dysbiosis impairs the structural and functional integrity of the ENS, leading to gut dysmotility in juvenile mice. EXPERIMENTAL APPROACH To induce gut dysbiosis, broad-spectrum antibiotics were administered by gavage to juvenile (3weeks old) male C57Bl/6 mice for 14 days. Bile acid composition in the intestinal lumen was analysed by liquid chromatography-mass spectrometry. Changes in intestinal motility were evaluated by stool frequency, transit of a fluorescent-labelled marker and isometric muscle responses of ileal full-thickness preparations to receptor and non-receptor-mediated stimuli. Alterations in ENS integrity were assessed by immunohistochemistry and Western blot analysis. KEY RESULTS Antibiotic treatment altered gastrointestinal transit, luminal bile acid metabolism and bowel architecture. Gut dysbiosis resulted in distorted glial network, loss of myenteric plexus neurons, altered cholinergic, tachykininergic and nitrergic neurotransmission associated with reduced number of nNOS neurons and different ileal distribution of the toll-like receptor TLR2. Functional defects were partly reversed by activation of TLR2 signalling. CONCLUSIONS AND IMPLICATIONS Gut dysbiosis caused complex morpho-functional neuromuscular rearrangements, characterized by structural defects of the ENS and increased tachykininergic neurotransmission. Altogether, our findings support the beneficial role of enteric microbiota for ENS homeostasis instrumental in ensuring proper gut neuromuscular function during critical stages of development.
Collapse
Affiliation(s)
- Valentina Caputi
- Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaPadovaItaly
| | - Ilaria Marsilio
- Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaPadovaItaly
| | - Viviana Filpa
- Department of Medicine and SurgeryUniversity of InsubriaVareseItaly
| | - Silvia Cerantola
- Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaPadovaItaly
- San Camillo HospitalTrevisoItaly
| | - Genny Orso
- IRCCS ‘E. Medea’ Bosisio PariniLeccoItaly
| | | | - Nicola Paccagnella
- Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaPadovaItaly
| | - Sara De Martin
- Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaPadovaItaly
| | - Monica Montopoli
- Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaPadovaItaly
| | - Stefano Dall'Acqua
- Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaPadovaItaly
| | - Francesca Crema
- Department of Internal Medicine and Therapeutics, Section of PharmacologyUniversity of PaviaPaviaItaly
| | | | | | | | - Sara Bogialli
- Department of Chemical SciencesUniversity of PadovaPadovaItaly
| | - Massimo Rugge
- Department of MedicineUniversity of PadovaPadovaItaly
| | - Patrizia Debetto
- Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaPadovaItaly
| | - Cristina Giaroni
- Department of Medicine and SurgeryUniversity of InsubriaVareseItaly
| | - Maria Cecilia Giron
- Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaPadovaItaly
| |
Collapse
|
32
|
Shapiro H, Suez J, Elinav E. Personalized microbiome-based approaches to metabolic syndrome management and prevention. J Diabetes 2017; 9:226-236. [PMID: 27787945 DOI: 10.1111/1753-0407.12501] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 10/08/2016] [Accepted: 10/24/2016] [Indexed: 12/11/2022] Open
Abstract
Personalized or precision medicine is a novel clinical approach targeted to the individual patient and based on integration of clinical, genetic, and environmental factors that define a patient uniquely from other individuals featuring similar clinical symptoms. Such a personalized medicine approach is increasingly applied for diagnosis, clinical stratification, and treatment of metabolic syndrome (MetS)-associated risks and diseases, including obesity, type 2 diabetes, non-alcoholic fatty liver disease, and their complications. One emerging factor that governs MetS manifestations is the microbiome, the composition, function, growth dynamics, associated metabolite profile and diverse effects of which on host immune and metabolic systems can all significantly affect metabolic homeostasis. Interindividual differences in microbiome composition and function, as well as personal variations in microbial-derived products, pave the way towards microbiome-based personalized medicine in treating MetS-related diseases.
Collapse
Affiliation(s)
- Hagit Shapiro
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Jotham Suez
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Elinav
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
33
|
Chand D, Avinash VS, Yadav Y, Pundle AV, Suresh CG, Ramasamy S. Molecular features of bile salt hydrolases and relevance in human health. Biochim Biophys Acta Gen Subj 2017; 1861:2981-2991. [DOI: 10.1016/j.bbagen.2016.09.024] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 09/20/2016] [Accepted: 09/23/2016] [Indexed: 01/18/2023]
|
34
|
Gonzalez FJ, Jiang C, Patterson AD. An Intestinal Microbiota-Farnesoid X Receptor Axis Modulates Metabolic Disease. Gastroenterology 2016; 151:845-859. [PMID: 27639801 PMCID: PMC5159222 DOI: 10.1053/j.gastro.2016.08.057] [Citation(s) in RCA: 268] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 08/02/2016] [Accepted: 08/12/2016] [Indexed: 02/07/2023]
Abstract
The gut microbiota is associated with metabolic diseases including obesity, insulin resistance, and nonalcoholic fatty liver disease, as shown by correlative studies and by transplant of microbiota from obese humans and mice into germ-free mice. Modification of the microbiota by treatment of high-fat diet (HFD)-fed mice with tempol or antibiotics resulted in decreased adverse metabolic phenotypes. This was owing to lower levels of the genera Lactobacillus and decreased bile salt hydrolase (BSH) activity. The decreased BSH resulted in increased levels of tauro-β-muricholic acid (MCA), a substrate of BSH and a potent farnesoid X receptor (FXR) antagonist. Mice lacking expression of FXR in the intestine were resistant to HFD-induced obesity, insulin resistance, and nonalcoholic fatty liver disease, thus confirming that intestinal FXR is involved in the potentiation of metabolic disease. A potent intestinal FXR antagonist, glycine-β-MCA (Gly-MCA), which is resistant to BSH, was developed, which, when administered to HFD-treated mice, mimics the effect of the altered microbiota on HFD-induced metabolic disease. Gly-MCA had similar effects on genetically obese leptin-deficient mice. The decrease in adverse metabolic phenotype by tempol, antibiotics, and Gly-MCA was caused by decreased serum ceramides. Mice lacking FXR in the intestine also have lower serum ceramide levels, and are resistant to HFD-induced metabolic disease, and this was reversed by injection of C16:0 ceramide. In mouse ileum, because of the presence of endogenous FXR agonists produced in the liver, FXR target genes involved in ceramide synthesis are activated and when Gly-MCA is administered they are repressed, which likely accounts for the decrease in serum ceramides. These studies show that ceramides produced in the ileum under control of FXR influence metabolic diseases.
Collapse
Affiliation(s)
- Frank J. Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, and the Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, P. R. China
| | - 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
| |
Collapse
|
35
|
Harakeh SM, Khan I, Kumosani T, Barbour E, Almasaudi SB, Bahijri SM, Alfadul SM, Ajabnoor GMA, Azhar EI. Gut Microbiota: A Contributing Factor to Obesity. Front Cell Infect Microbiol 2016; 6:95. [PMID: 27625997 PMCID: PMC5003832 DOI: 10.3389/fcimb.2016.00095] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 08/17/2016] [Indexed: 12/25/2022] Open
Abstract
Obesity, a global epidemic of the modern era, is a risk factor for cardiovascular diseases (CVD) and diabetes. The pervasiveness of obesity and overweight in both developed as well as developing populations is on the rise and placing a huge burden on health and economic resources. Consequently, research to control this emerging epidemic is of utmost importance. Recently, host interactions with their resident gut microbiota (GM) have been reported to be involved in the pathogenesis of many metabolic diseases, including obesity, diabetes, and CVD. Around 10(14) microorganisms reside within the lower human intestine and many of these 10(14) microorganisms have developed mutualistic or commensal associations with the host and actively involved in many physiological processes of the host. However, dysbiosis (altered gut microbial composition) with other predisposing genetic and environmental factors, may contribute to host metabolic disorders resulting in many ailments. Therefore, delineating the role of GM as a contributing factor to obesity is the main objective of this review. Obesity research, as a field is expanding rapidly due to major advances in nutrigenomics, metabolomics, RNA silencing, epigenetics, and other disciplines that may result in the emergence of new technologies and methods to better interpret causal relationships between microbiota and obesity.
Collapse
Affiliation(s)
- Steve M Harakeh
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University Jeddah, Saudi Arabia
| | - Imran Khan
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz UniversityJeddah, Saudi Arabia; Department of Biochemistry, Faculty of Science, King Abdulaziz UniversityJeddah, Saudi Arabia
| | - Taha Kumosani
- Department of Biochemistry, Faculty of Science, King Abdulaziz UniversityJeddah, Saudi Arabia; Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz UniversityJeddah, Saudi Arabia
| | - Elie Barbour
- Department of Animal and Veterinary Sciences, Faculty of Agricultural and Food Sciences, American University of BeirutBeirut, Lebanon; Department of Biochemistry, Faculty of Science, King Abdulaziz UniversityJeddah, Saudi Arabia
| | - Saad B Almasaudi
- Biology Department, Faculty of Science, King Abdulaziz University Jeddah, Saudi Arabia
| | - Suhad M Bahijri
- Clinical Biochemistry Department, College of Medicine, Nutrition Unit-King Fahd Medical Research Center, King Abdulaziz University Jeddah, Saudi Arabia
| | | | - Ghada M A Ajabnoor
- Clinical Biochemistry Department, College of Medicine, Nutrition Unit-King Fahd Medical Research Center, King Abdulaziz University Jeddah, Saudi Arabia
| | - Esam I Azhar
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz UniversityJeddah, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Science, King Abdulaziz UniversityJeddah, Saudi Arabia
| |
Collapse
|
36
|
Bashiardes S, Shapiro H, Rozin S, Shibolet O, Elinav E. Non-alcoholic fatty liver and the gut microbiota. Mol Metab 2016; 5:782-94. [PMID: 27617201 PMCID: PMC5004228 DOI: 10.1016/j.molmet.2016.06.003] [Citation(s) in RCA: 159] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 06/04/2016] [Accepted: 06/08/2016] [Indexed: 02/06/2023] Open
Abstract
Background Non-alcoholic fatty liver (NAFLD) is a common, multi-factorial, and poorly understood liver disease whose incidence is globally rising. NAFLD is generally asymptomatic and associated with other manifestations of the metabolic syndrome. Yet, up to 25% of NAFLD patients develop a progressive inflammatory liver disease termed non-alcoholic steatohepatitis (NASH) that may progress towards cirrhosis, hepatocellular carcinoma, and the need for liver transplantation. In recent years, several lines of evidence suggest that the gut microbiome represents a significant environmental factor contributing to NAFLD development and its progression into NASH. Suggested microbiome-associated mechanisms contributing to NAFLD and NASH include dysbiosis-induced deregulation of the gut endothelial barrier function, which facilitates systemic bacterial translocation, and intestinal and hepatic inflammation. Furthermore, increased microbiome-modulated metabolites such as lipopolysaccharides, short chain fatty acids (SCFAs), bile acids, and ethanol, may affect liver pathology through multiple direct and indirect mechanisms. Scope of review Herein, we discuss the associations, mechanisms, and clinical implications of the microbiome's contribution to NAFLD and NASH. Understanding these contributions to the development of fatty liver pathogenesis and its clinical course may serve as a basis for development of therapeutic microbiome-targeting approaches for treatment and prevention of NAFLD and NASH. Major conclusions Intestinal host–microbiome interactions play diverse roles in the pathogenesis and progression of NAFLD and NASH. Elucidation of the mechanisms driving these microbial effects on the pathogenesis of NAFLD and NASH may enable to identify new diagnostic and therapeutic targets of these common metabolic liver diseases. This article is part of a special issue on microbiota.
Collapse
Affiliation(s)
- Stavros Bashiardes
- Immunology Department, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Hagit Shapiro
- Immunology Department, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Shachar Rozin
- Immunology Department, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Oren Shibolet
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel; Department of Gastroenterology, Tel Aviv Medical Center, Tel Aviv 6423906, Israel
| | - Eran Elinav
- Immunology Department, Weizmann Institute of Science, 76100 Rehovot, Israel
| |
Collapse
|
37
|
Obeticholic acid reduces bacterial translocation and inhibits intestinal inflammation in cirrhotic rats. J Hepatol 2016; 64:1049-1057. [PMID: 26723896 DOI: 10.1016/j.jhep.2015.12.010] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 11/22/2015] [Accepted: 12/18/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS In advanced cirrhosis, gut bacterial translocation is the consequence of intestinal barrier disruption and leads to bacterial infection. Bile acid abnormalities in cirrhosis could play a role in the integrity of the intestinal barrier and the control of microbiota, mainly through the farnesoid X receptor. We investigated the long-term effects of the farnesoid X receptor agonist, obeticholic acid, on gut bacterial translocation, intestinal microbiota composition, barrier integrity and inflammation in rats with CCl4-induced cirrhosis with ascites. METHODS Cirrhotic rats received a 2-week course of obeticholic acid or vehicle starting once ascites developed. We then determined: bacterial translocation by mesenteric lymph node culture, ileum expression of antimicrobial peptides and tight junction proteins by qPCR, fecal albumin loss, enteric bacterial load and microbiota composition by qPCR and pyrosequencing of ileum mucosa-attached contents, and intestinal inflammation by cytometry of the inflammatory infiltrate. RESULTS Obeticholic acid reduced bacterial translocation from 78.3% to 33.3% (p<0.01) and upregulated the expression of the farnesoid X receptor-associated gene small heterodimer partner. Treatment improved ileum expression of antimicrobial peptides, angiogenin-1 and alpha-5-defensin, tight junction proteins zonulin-1 and occludin, and reduced fecal albumin loss and liver fibrosis. Enteric bacterial load normalized, and the distinctive mucosal microbiota of cirrhosis was reduced. Gut immune cell infiltration was reduced and inflammatory cytokine and Toll-like receptor 4 expression normalized. CONCLUSIONS In ascitic cirrhotic rats, obeticholic acid reduces gut bacterial translocation via several complementary mechanisms at the intestinal level. This agent could be used as an alternative to antibiotics to prevent bacterial infection in cirrhosis.
Collapse
|
38
|
Role of the microbiome in the normal and aberrant glycemic response. CLINICAL NUTRITION EXPERIMENTAL 2016. [DOI: 10.1016/j.yclnex.2016.01.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
39
|
Penney NC, Kinross J, Newton RC, Purkayastha S. The role of bile acids in reducing the metabolic complications of obesity after bariatric surgery: a systematic review. Int J Obes (Lond) 2015; 39:1565-74. [PMID: 26081915 DOI: 10.1038/ijo.2015.115] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 03/05/2015] [Accepted: 05/31/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND Bariatric surgery is currently the most efficacious treatment for obesity and its associated metabolic co-morbidities, such as diabetes. The metabolic improvements occur through both weight-dependent and weight-independent mechanisms. Bile acids (BAs) have emerged as key signalling molecules that have a central role in modulating many of the physiological effects seen after bariatric surgery. This systematic review assesses the evidence from both human and animal studies for the role of BAs in reducing the metabolic complications of obesity following bariatric surgery. METHODS We conducted a systematic search of Medline and Embase databases to identify all articles investigating the role of BAs in mediating the metabolic changes observed following bariatric surgery in both animal and human studies. Boolean logic was used with relevant search terms, including the following MeSH terms: 'bile acids and salts', 'bariatric surgery', 'metabolic surgery', 'gastrointestinal tract/surgery' and 'obesity/surgery'. RESULTS Following database searches (n=1197), inclusion from bibliography searches (n=2) and de-duplication (n=197), 1002 search results were returned. Of these, 132 articles were selected for full-text review, of which 38 articles were deemed relevant and included in the review. The findings support the effects of BAs on satiety, lipid and cholesterol metabolism, incretins and glucose homoeostasis, energy metabolism, gut microbiota and endoplasmic reticulum stress following bariatric surgery. Many of these metabolic effects are modulated through the BA receptors FXR and TGR5. We also explore a possible link between BAs and carcinogenesis following bariatric surgery. CONCLUSIONS Overall there is good evidence to support the role of BAs in the metabolic effects of bariatric surgery through the above mechanisms. BAs could serve as a novel therapeutic pharmacological target for the treatment of obesity and its associated co-morbidities.
Collapse
Affiliation(s)
- N C Penney
- Department of Surgery and Cancer, St Mary's Hospital, Imperial College London, London, UK
| | - J Kinross
- Department of Surgery and Cancer, St Mary's Hospital, Imperial College London, London, UK
| | - R C Newton
- Department of Surgery and Cancer, St Mary's Hospital, Imperial College London, London, UK
| | - S Purkayastha
- Department of Surgery and Cancer, St Mary's Hospital, Imperial College London, London, UK
| |
Collapse
|
40
|
Martoni CJ, Labbé A, Ganopolsky JG, Prakash S, Jones ML. Changes in bile acids, FGF-19 and sterol absorption in response to bile salt hydrolase active L. reuteri NCIMB 30242. Gut Microbes 2015; 6:57-65. [PMID: 25612224 PMCID: PMC4615650 DOI: 10.1080/19490976.2015.1005474] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 12/03/2014] [Accepted: 01/05/2015] [Indexed: 02/07/2023] Open
Abstract
The size and composition of the circulating bile acid (BA) pool are important factors in regulating the human gut microbiota. Disrupted regulation of BA metabolism is implicated in several chronic diseases. Bile salt hydrolase (BSH)-active Lactobacillus reuteri NCIMB 30242, previously shown to decrease LDL-cholesterol and increase circulating BA, was investigated for its dose response effect on BA profile in a pilot clinical study. Ten otherwise healthy hypercholesterolemic adults, recruited from a clinical trial site in London, ON, were randomized to consume delayed release or standard release capsules containing L. reuteri NCIMB 30242 in escalating dose over 4 weeks. In another aspect, 4 healthy normocholesterolemic subjects with LDL-C below 3.4 mmol/l received delayed release L. reuteri NCIMB 30242 at a constant dose over 4 weeks. The primary outcome measure was the change in plasma BA profile over the intervention period. Additional outcomes included circulating fibroblast growth factor (FGF)-19, plant sterols and LDL-cholesterol as well as fecal microbiota and bsh gene presence. After one week of intervention subjects receiving delayed release L. reuteri NCIMB 30242 increased total BA by 1.13 ± 0.67 μmol/l (P = 0.02), conjugated BA by 0.67 ± 0.39 μmol/l (P = 0.02) and unconjugated BA by 0.46 ± 0.43 μmol/l (P = 0.07), which represented a greater than 2-fold change relative to baseline. Increases in BA were largely maintained post-week 1 and were generally correlated with FGF-19 and inversely correlated with plant sterols. This is the first clinical support showing that a BSH-active probiotic can significantly and rapidly influence BA metabolism and may prove useful in chronic diseases beyond hypercholesterolemia.
Collapse
Affiliation(s)
| | | | | | - Satya Prakash
- Biomedical Technology and Cell Therapy Research Laboratory; Dept. of Biomedical Engineering; Faculty of Medicine; McGill University; Montreal, QC Canada
- Micropharma Limited; Montreal, QC Canada
| | - Mitchell L Jones
- Biomedical Technology and Cell Therapy Research Laboratory; Dept. of Biomedical Engineering; Faculty of Medicine; McGill University; Montreal, QC Canada
- Micropharma Limited; Montreal, QC Canada
| |
Collapse
|
41
|
Moran CP, Shanahan F. Gut microbiota and obesity: role in aetiology and potential therapeutic target. Best Pract Res Clin Gastroenterol 2014; 28:585-97. [PMID: 25194177 DOI: 10.1016/j.bpg.2014.07.005] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Revised: 06/20/2014] [Accepted: 07/05/2014] [Indexed: 01/31/2023]
Abstract
Obesity is epidemic; chronic energy surplus is clearly important in obesity development but other factors are at play. Indigenous gut microbiota are implicated in the aetiopathogenesis of obesity and obesity-related disorders. Evidence from murine models initially suggested a role for the gut microbiota in weight regulation and the microbiota has been shown to contribute to the low grade inflammation that characterises obesity. The microbiota and its metabolites mediate some of the alterations of the microbiota-gut-brain axis, the endocannabinoid system, and bile acid metabolism, found in obesity-related disorders. Modulation of the gut microbiota is an attractive proposition for prevention or treatment of obesity, particularly as traditional measures have been sub-optimal.
Collapse
Affiliation(s)
- Carthage P Moran
- Dept. Medicine and Alimentary Pharmabiotic Centre, University College Cork, National University of Ireland, Cork, Ireland
| | - Fergus Shanahan
- Dept. Medicine and Alimentary Pharmabiotic Centre, University College Cork, National University of Ireland, Cork, Ireland.
| |
Collapse
|
42
|
Shapiro H, Thaiss CA, Levy M, Elinav E. The cross talk between microbiota and the immune system: metabolites take center stage. Curr Opin Immunol 2014; 30:54-62. [PMID: 25064714 DOI: 10.1016/j.coi.2014.07.003] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 06/28/2014] [Accepted: 07/06/2014] [Indexed: 02/07/2023]
Abstract
The human meta-organism consists of more than 90% of microbial cells. The gastrointestinal tract harbors trillions of commensal microorganisms that influence the development and homeostasis of the host. Alterations in composition and function of the microbiota, termed dysbiosis, have been implicated in a multitude of metabolic and inflammatory diseases in humans. Thus, understanding the molecular underpinnings the cross talk between commensal bacteria and their host during homeostasis and dysbiosis may hold the key to understanding many idiopathic diseases. While most attention has focused on the innate recognition of immune-stimulatory bacterial molecules, such as cell wall components and nucleic acids, we emphasize here the impact of diet-dependent microbial metabolites on the development and function of the immune system.
Collapse
Affiliation(s)
- Hagit Shapiro
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Christoph A Thaiss
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Maayan Levy
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Elinav
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
43
|
Tsuei J, Chau T, Mills D, Wan YJY. Bile acid dysregulation, gut dysbiosis, and gastrointestinal cancer. Exp Biol Med (Maywood) 2014; 239:1489-504. [PMID: 24951470 DOI: 10.1177/1535370214538743] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Because of increasingly widespread sedentary lifestyles and diets high in fat and sugar, the global diabetes and obesity epidemic continues to grow unabated. A substantial body of evidence has been accumulated which associates diabetes and obesity to dramatically higher risk of cancer development, particularly in the liver and gastrointestinal tract. Additionally, diabetic and obese individuals have been shown to suffer from dysregulation of bile acid (BA) homeostasis and dysbiosis of the intestinal microbiome. Abnormally elevated levels of cytotoxic secondary BAs and a pro-inflammatory shift in gut microbial profile have individually been linked to numerous enterohepatic diseases including cancer. However, recent findings have implicated a detrimental interplay between BA dysregulation and intestinal dysbiosis that promotes carcinogenesis along the gut-liver axis. This review seeks to examine the currently investigated interactions between the regulation of BA metabolism and activity of the intestinal microbiota and how these interactions can drive cancer formation in the context of diabesity. The precarcinogenic effects of BA dysregulation and gut dysbiosis including excessive inflammation, heightened oxidative DNA damage, and increased cell proliferation are discussed. Furthermore, by focusing on the mediatory roles of BA nuclear receptor farnesoid x receptor, ileal transporter apical sodium dependent BA transporter, and G-coupled protein receptor TGR5, this review attempts to connect BA dysregulation, gut dysbiosis, and enterohepatic carcinogenesis at a mechanistic level. A better understanding of the intricate interplay between BA homeostasis and gut microbiome can yield novel avenues to combat the impending rise in diabesity-related cancers.
Collapse
Affiliation(s)
- Jessica Tsuei
- Department of Pathology and Laboratory Medicine, University of California at Davis Medical Center, Sacramento, CA 95831, USA
| | - Thinh Chau
- Department of Pathology and Laboratory Medicine, University of California at Davis Medical Center, Sacramento, CA 95831, USA
| | - David Mills
- Department of Food Science and Technology, Department of Viticulture and Enology, Foods for Health Institute, University of California, Davis, CA 95616, USA
| | - Yu-Jui Yvonne Wan
- Department of Pathology and Laboratory Medicine, University of California at Davis Medical Center, Sacramento, CA 95831, USA
| |
Collapse
|
44
|
Zhang Y, Limaye PB, Renaud HJ, Klaassen CD. Effect of various antibiotics on modulation of intestinal microbiota and bile acid profile in mice. Toxicol Appl Pharmacol 2014; 277:138-45. [PMID: 24657338 DOI: 10.1016/j.taap.2014.03.009] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 03/10/2014] [Accepted: 03/12/2014] [Indexed: 12/24/2022]
Abstract
Antibiotic treatments have been used to modulate intestinal bacteria and investigate the role of intestinal bacteria on bile acid (BA) homeostasis. However, knowledge on which intestinal bacteria and bile acids are modified by antibiotics is limited. In the present study, mice were administered various antibiotics, 47 of the most abundant bacterial species in intestine, as well as individual BAs in plasma, liver, and intestine were quantified. Compared to the two antibiotic combinations (vancomycin+imipenem and cephalothin+neomycin), the three single antibiotics (metronidazole, ciprofloxacin and aztreonam) have less effect on intestinal bacterial profiles, and thus on host BA profiles and mRNA expression of genes that are important for BA homeostasis. The two antibiotic combinations decreased the ratio of Firmicutes to Bacteroidetes in intestine, as well as most secondary BAs in serum, liver and intestine. Additionally, the two antibiotic combinations significantly increased mRNA of the hepatic BA uptake transporters (Ntcp and Oatp1b2) and canalicular BA efflux transporters (Bsep and Mrp2), but decreased mRNA of the hepatic BA synthetic enzyme Cyp8b1, suggesting an elevated enterohepatic circulation of BAs. Interestingly, the two antibiotic combinations tended to have opposite effect on the mRNAs of most intestinal genes, which tended to be inhibited by vancomycin+imipenem but stimulated by cephalothin+neomycin. To conclude, the present study clearly shows that various antibiotics have distinct effects on modulating intestinal bacteria and host BA metabolism.
Collapse
Affiliation(s)
- Youcai Zhang
- Department of Internal Medicine, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| | - Pallavi B Limaye
- Department of Internal Medicine, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| | - Helen J Renaud
- Department of Internal Medicine, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| | - Curtis D Klaassen
- Department of Internal Medicine, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA.
| |
Collapse
|
45
|
Jones ML, Martoni CJ, Ganopolsky JG, Labbé A, Prakash S. The human microbiome and bile acid metabolism: dysbiosis, dysmetabolism, disease and intervention. Expert Opin Biol Ther 2014; 14:467-82. [PMID: 24479734 DOI: 10.1517/14712598.2014.880420] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Recent evidence indicates that the human gut microbiome plays a significant role in health and disease. Dysbiosis, defined as a pathological imbalance in a microbial community, is becoming increasingly appreciated as a 'central environmental factor' that is both associated with complex phenotypes and affected by host genetics, diet and antibiotic use. More recently, a link has been established between the dysmetabolism of bile acids (BAs) in the gut to dysbiosis. AREAS COVERED BAs, which are transformed by the gut microbiota, have been shown to regulate intestinal homeostasis and are recognized as signaling molecules in a wide range of metabolic processes. This review will examine the connection between BA metabolism as it relates to the gut microbiome and its implication in health and disease. EXPERT OPINION A disrupted gut microbiome, including a reduction of bile salt hydrolase (BSH)-active bacteria, can significantly impair the metabolism of BAs and may result in an inability to maintain glucose homeostasis as well as normal cholesterol breakdown and excretion. To better understand the link between dysbiosis, BA dysmetabolism and chronic degenerative disease, large-scale metagenomic sequencing studies, metatranscriptomics, metaproteomics and metabolomics should continue to catalog functional diversity in the gastrointestinal tract of both healthy and diseased populations. Further, BSH-active probiotics should continue to be explored as treatment options to help restore metabolic levels.
Collapse
Affiliation(s)
- Mitchell L Jones
- McGill University, Artificial Cells and Organs Research Centre, Department of Biomedical Engineering and Physiology, Biomedical Technology and Cell Therapy Research Laboratory, Faculty of Medicine , 3775 University Street, Montreal, Quebec, H3A2B4 , Canada +1 514 398 3676 ; +1 514 398 7461 ;
| | | | | | | | | |
Collapse
|
46
|
Pan D, Yu Z. Intestinal microbiome of poultry and its interaction with host and diet. Gut Microbes 2014; 5:108-19. [PMID: 24256702 PMCID: PMC4049927 DOI: 10.4161/gmic.26945] [Citation(s) in RCA: 509] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 10/23/2013] [Accepted: 10/24/2013] [Indexed: 02/03/2023] Open
Abstract
The gastrointestinal (GI) tract of poultry is densely populated with microorganisms which closely and intensively interact with the host and ingested feed. The gut microbiome benefits the host by providing nutrients from otherwise poorly utilized dietary substrates and modulating the development and function of the digestive and immune system. In return, the host provides a permissive habitat and nutrients for bacterial colonization and growth. Gut microbiome can be affected by diet, and different dietary interventions are used by poultry producers to enhance bird growth and reduce risk of enteric infection by pathogens. There also exist extensive interactions among members of the gut microbiome. A comprehensive understanding of these interactions will help develop new dietary or managerial interventions that can enhance bird growth, maximize host feed utilization, and protect birds from enteric diseases caused by pathogenic bacteria.
Collapse
Affiliation(s)
- Deng Pan
- Department of Animal Sciences; The Ohio State University; Columbus, OH USA
| | - Zhongtang Yu
- Department of Animal Sciences; The Ohio State University; Columbus, OH USA
| |
Collapse
|
47
|
Ridaura VK, Faith JJ, Rey FE, Cheng J, Duncan AE, Kau AL, Griffin NW, Lombard V, Henrissat B, Bain JR, Muehlbauer MJ, Ilkayeva O, Semenkovich CF, Funai K, Hayashi DK, Lyle BJ, Martini MC, Ursell LK, Clemente JC, Van Treuren W, Walters WA, Knight R, Newgard CB, Heath AC, Gordon JI. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science 2013; 341:1241214. [PMID: 24009397 DOI: 10.1126/science.1241214] [Citation(s) in RCA: 2695] [Impact Index Per Article: 224.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The role of specific gut microbes in shaping body composition remains unclear. We transplanted fecal microbiota from adult female twin pairs discordant for obesity into germ-free mice fed low-fat mouse chow, as well as diets representing different levels of saturated fat and fruit and vegetable consumption typical of the U.S. diet. Increased total body and fat mass, as well as obesity-associated metabolic phenotypes, were transmissible with uncultured fecal communities and with their corresponding fecal bacterial culture collections. Cohousing mice harboring an obese twin's microbiota (Ob) with mice containing the lean co-twin's microbiota (Ln) prevented the development of increased body mass and obesity-associated metabolic phenotypes in Ob cage mates. Rescue correlated with invasion of specific members of Bacteroidetes from the Ln microbiota into Ob microbiota and was diet-dependent. These findings reveal transmissible, rapid, and modifiable effects of diet-by-microbiota interactions.
Collapse
Affiliation(s)
- Vanessa K Ridaura
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63108, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Differential activation of the human farnesoid X receptor depends on the pattern of expressed isoforms and the bile acid pool composition. Biochem Pharmacol 2013; 86:926-39. [PMID: 23928191 DOI: 10.1016/j.bcp.2013.07.022] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2013] [Revised: 07/23/2013] [Accepted: 07/23/2013] [Indexed: 12/15/2022]
Abstract
The farnesoid X receptor (FXR) is a key sensor in bile acid homeostasis. Although four human FXR isoforms have been identified, the physiological role of this diversity is poorly understood. Here we investigated their subcellular localization, agonist sensitivity and response of target genes. Measurement of mRNA revealed that liver predominantly expressed FXRα1(+/-), whereas FXRα2(+/-) were the most abundant isoforms in kidney and intestine. In all cases, the proportion of FXRα(1/2)(+) and FXRα(1/2)(-) isoforms, i.e., with and without a 12bp insert, respectively, was approximately 50%. When FXR was expressed in liver and intestinal cells the magnitude of the response to GW4064 and bile acids differs among FXR isoforms. In both cell types the strongest response was that of FXRα1(-). Different efficacy of bile acids species to activate FXR was found. The four FXR isoforms shared the order of sensitivity to bile acids species. When in FXR-deficient cells FXR was transfected, unconjugated, but not taurine- and glycine-amidated bile acids, were able to activate FXR. In contrast, human hepatocytes and cell lines showing an endogenous expression of FXR were sensitive to both unconjugated and conjugated bile acids. This suggests that to activate FXR conjugated, but not unconjugated, bile acids require additional component(s) of the intracellular machinery not related with uptake processes, which are missing in some tumor cells. In conclusion, cell-specific pattern of FXR isoforms determine the overall tissue sensitivity to FXR agonists and may be involved in the differential response of FXR target genes to FXR activation.
Collapse
|
49
|
Miyata M, Yamakawa H, Hayashi K, Kuribayashi H, Yamazoe Y, Yoshinari K. Ileal apical sodium-dependent bile acid transporter protein levels are down-regulated through ubiquitin-dependent protein degradation induced by bile acids. Eur J Pharmacol 2013; 714:507-14. [PMID: 23872411 DOI: 10.1016/j.ejphar.2013.06.036] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 06/15/2013] [Accepted: 06/21/2013] [Indexed: 12/15/2022]
Abstract
The ileal apical sodium-dependent bile acid transporter (ASBT or SLC10A2) has a crucial role in intestinal bile acid absorption. We previously reported that enterobacteria-mediated bile acid conversion was involved in the alteration of ileal ASBT expression levels. In the present study, to investigate the hypothesis that ileal ASBT protein levels are post-translationally regulated by enterobacteria-associated bile acids, alteration of ileal ASBT protein levels was analysed in mice 12 h and 24 h after anti-bacterial drug ampicillin (ABPC) treatment (100 mg/kg, single shot) that altered bile acid composition in the intestinal lumen. In ABPC-treated mice, enterobacteria-biotransformed bile acid, taurodeoxycholic acid (TDCA) and cholic acid (CA) levels were decreased, whereas taurocholic acid (TCA) and tauro-β-muricholic acid levels were increased in the intestinal lumen. Ileal ASBT protein levels in brush-border membrane vesicles (BBMVs), but not ileal Asbt mRNA levels, were significantly increased in the ABPC-treated mice, and the extent of ubiquitination of the ileal ASBT protein was reduced in the ABPC-treated mice. Treatment of ABPC-pretreated mice with CA or TDCA, but not TCA, significantly decreased ileal ASBT protein levels and increased the extent of ubiquitination of ileal ASBT protein. Treatment of mice with the lysosome inhibitor, chloroquine, or the proteasome inhibitor, MG132, increased ileal ASBT protein levels in BBMVs. CA-mediated reduction of ASBT protein levels in the ABPC-pretreated mice was attenuated by co-treatment with chloroquine or MG132. These results suggest that ileal ASBT protein is degraded by a ubiquitin-dependent pathway in response to enterobacteria-associated bile acids.
Collapse
Affiliation(s)
- Masaaki Miyata
- Division of Drug Metabolism and Molecular Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan.
| | | | | | | | | | | |
Collapse
|
50
|
Sayin SI, Wahlström A, Felin J, Jäntti S, Marschall HU, Bamberg K, Angelin B, Hyötyläinen T, Orešič M, Bäckhed F. Gut microbiota regulates bile acid metabolism by reducing the levels of tauro-beta-muricholic acid, a naturally occurring FXR antagonist. Cell Metab 2013; 17:225-35. [PMID: 23395169 DOI: 10.1016/j.cmet.2013.01.003] [Citation(s) in RCA: 1659] [Impact Index Per Article: 138.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2011] [Revised: 08/23/2012] [Accepted: 01/10/2013] [Indexed: 12/13/2022]
Abstract
Bile acids are synthesized from cholesterol in the liver and further metabolized by the gut microbiota into secondary bile acids. Bile acid synthesis is under negative feedback control through activation of the nuclear receptor farnesoid X receptor (FXR) in the ileum and liver. Here we profiled the bile acid composition throughout the enterohepatic system in germ-free (GF) and conventionally raised (CONV-R) mice. We confirmed a dramatic reduction in muricholic acid, but not cholic acid, levels in CONV-R mice. Rederivation of Fxr-deficient mice as GF demonstrated that the gut microbiota regulated expression of fibroblast growth factor 15 in the ileum and cholesterol 7α-hydroxylase (CYP7A1) in the liver by FXR-dependent mechanisms. Importantly, we identified tauro-conjugated beta- and alpha-muricholic acids as FXR antagonists. These studies suggest that the gut microbiota not only regulates secondary bile acid metabolism but also inhibits bile acid synthesis in the liver by alleviating FXR inhibition in the ileum.
Collapse
Affiliation(s)
- Sama I Sayin
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine and Sahlgrenska Center for Cardiovascular and Metabolic Research, University of Gothenburg, 413 45 Gothenburg, Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|