801
|
Jadhav K, Xu Y, Xu Y, Li Y, Xu J, Zhu Y, Adorini L, Lee YK, Kasumov T, Yin L, Zhang Y. Reversal of metabolic disorders by pharmacological activation of bile acid receptors TGR5 and FXR. Mol Metab 2018; 9:131-140. [PMID: 29361497 PMCID: PMC5870099 DOI: 10.1016/j.molmet.2018.01.005] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 12/25/2017] [Accepted: 01/08/2018] [Indexed: 02/08/2023] Open
Abstract
Objectives Activation of the bile acid (BA) receptors farnesoid X receptor (FXR) or G protein-coupled bile acid receptor (GPBAR1; TGR5) improves metabolic homeostasis. In this study, we aim to determine the impact of pharmacological activation of bile acid receptors by INT-767 on reversal of diet-induced metabolic disorders, and the relative contribution of FXR vs. TGR5 to INT-767's effects on metabolic parameters. Methods Wild-type (WT), Tgr5−/−, Fxr−/−, Apoe−/− and Shp−/− mice were used to investigate whether and how BA receptor activation by INT-767, a semisynthetic agonist for both FXR and TGR5, could reverse diet-induced metabolic disorders. Results INT-767 reversed HFD-induced obesity dependent on activation of both TGR5 and FXR and also reversed the development of atherosclerosis and non-alcoholic fatty liver disease (NAFLD). Mechanistically, INT-767 improved hypercholesterolemia by activation of FXR and induced thermogenic genes via activation of TGR5 and/or FXR. Furthermore, INT-767 inhibited several lipogenic genes and de novo lipogenesis in the liver via activation of FXR. We identified peroxisome proliferation-activated receptor γ (PPARγ) and CCAAT/enhancer-binding protein α (CEBPα) as novel FXR-regulated genes. FXR inhibited PPARγ expression by inducing small heterodimer partner (SHP) whereas the inhibition of CEBPα by FXR was SHP-independent. Conclusions BA receptor activation can reverse obesity, NAFLD, and atherosclerosis by specific activation of FXR or TGR5. Our data suggest that, compared to activation of FXR or TGR5 only, dual activation of both FXR and TGR5 is a more attractive strategy for treatment of common metabolic disorders. Bile acid receptor activation improves lipid homeostasis primarily via activation of FXR. Bile acid receptor activation reverses DIO and induces energy expenditure. Bile acid receptor activation reverses diet-induced NAFLD and atherosclerosis. Bile acid receptor activation inhibits hepatic lipogenesis via activation of FXR. Activation of FXR inhibits PPARγ dependent on SHP and CEBPα independent of SHP.
Collapse
Affiliation(s)
- Kavita Jadhav
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44272, USA
| | - Yang Xu
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44272, USA
| | - Yanyong Xu
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44272, USA
| | - Yuanyuan Li
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44272, USA
| | - Jiesi Xu
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44272, USA; State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yingdong Zhu
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44272, USA
| | | | - Yoon Kwang Lee
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44272, USA
| | - Takhar Kasumov
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44272, USA
| | - Liya Yin
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44272, USA
| | - Yanqiao Zhang
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44272, USA.
| |
Collapse
|
802
|
Jena PK, Sheng L, Di Lucente J, Jin LW, Maezawa I, Wan YJY. Dysregulated bile acid synthesis and dysbiosis are implicated in Western diet-induced systemic inflammation, microglial activation, and reduced neuroplasticity. FASEB J 2018; 32:2866-2877. [PMID: 29401580 DOI: 10.1096/fj.201700984rr] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The goal of this study was to identify the intrinsic links that explain the effect of a Western diet (WD) on cognitive dysfunction. Specific pathogen-free, wild-type mice were fed either a control diet (CD) or a high-fat, high-sucrose WD after weaning and were euthanized at 10 mo of age to study the pathways that affect cognitive health. The results showed that long-term WD intake reduced hippocampal synaptic plasticity and the level of brain-derived neurotrophic factor mRNA in the brain and isolated microglia. A WD also activated ERK1/2 and reduced postsynaptic density-95 in the brain, suggesting postsynaptic damage. Moreover, WD-fed mice had increased inflammatory signaling in the brain, ileum, liver, adipose tissue, and spleen, which was accompanied by microglia activation. In the brain, as well as in the digestive tract, a WD reduced signaling regulated by retinoic acid and bile acids (BAs), whose receptors form heterodimers to control metabolism and inflammation. Furthermore, a WD intake caused dysbiosis and dysregulated BA synthesis with reduced endogenous ligands for BA receptors, i.e., farnesoid X receptor and G-protein-coupled bile acid receptor in the liver and brain. Together, dysregulated BA synthesis and dysbiosis were accompanied by systemic inflammation, microglial activation, and reduced neuroplasticity induced by WD.-Jena, P. K., Sheng, L., Di Lucente, J., Jin, L.-W., Maezawa, I., Wan, Y.-J. Y. Dysregulated bile acid synthesis and dysbiosis are implicated in Western diet-induced systemic inflammation, microglial activation, and reduced neuroplasticity.
Collapse
Affiliation(s)
- Prasant Kumar Jena
- Department of Medical Pathology and Laboratory Medicine University of California, Davis, Sacramento, California, USA; and
| | - Lili Sheng
- Department of Medical Pathology and Laboratory Medicine University of California, Davis, Sacramento, California, USA; and
| | - Jacopo Di Lucente
- Department of Medical Pathology and Laboratory Medicine University of California, Davis, Sacramento, California, USA; and.,Medical Investigation of Neurodevelopmental Disorders Institute, University of California, Davis, Sacramento, California, USA
| | - Lee-Way Jin
- Department of Medical Pathology and Laboratory Medicine University of California, Davis, Sacramento, California, USA; and.,Medical Investigation of Neurodevelopmental Disorders Institute, University of California, Davis, Sacramento, California, USA
| | - Izumi Maezawa
- Department of Medical Pathology and Laboratory Medicine University of California, Davis, Sacramento, California, USA; and.,Medical Investigation of Neurodevelopmental Disorders Institute, University of California, Davis, Sacramento, California, USA
| | - Yu-Jui Yvonne Wan
- Department of Medical Pathology and Laboratory Medicine University of California, Davis, Sacramento, California, USA; and
| |
Collapse
|
803
|
Zhang P, Meng X, Li D, Calderone R, Mao D, Sui B. Commensal Homeostasis of Gut Microbiota-Host for the Impact of Obesity. Front Physiol 2018; 8:1122. [PMID: 29358923 PMCID: PMC5766676 DOI: 10.3389/fphys.2017.01122] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 12/19/2017] [Indexed: 12/22/2022] Open
Abstract
Gut microbiota and their metabolites have been linked to a series of chronic diseases such as obesity and other metabolic dysfunctions. Obesity is an increasingly serious international health issue that may lead to a risk of insulin resistance and other metabolic diseases. The relationship between gut microbiota and the host is both interdependent and relatively independent. In this review, the causality of gut microbiota and its role in the pathogenesis and intervention of obesity is comprehensively presented to include human genotype, enterotypes, interactions of gut microbiota with the host, microbial metabolites, and energy homeostasis all of which may be influenced by dietary nutrition. Diet can enhance, inhibit, or even change the composition and functions of the gut microbiota. The metabolites they produce depend upon the dietary substrates provided, some of which have indispensable functions for the host. Therefore, diet is a key factor that maintains or not a healthy commensal relationship. In addition, the specific genotype of the host may impact the phylogenetic compositions of gut microbiota through the production of host metabolites. The commensal homeostasis of gut microbiota is favored by a balance of microbial composition, metabolites, and energy. Ultimately the desired commensal relationship is one of mutual support. This article analyzes the clues that result in patterns of commensal homeostasis. A deeper understanding of these interactions is beneficial for developing effective prevention, diagnosis, and personalized therapeutic strategies to combat obesity and other metabolic diseases. The idea we discuss is meant to improve human health by shaping or modulating the beneficial gut microbiota.
Collapse
Affiliation(s)
- Pengyi Zhang
- Sport Science Research Center, Shandong Sport University, Jinan, China.,Department of Microbiology & Immunology, Georgetown University Medical Center, Washington, DC, United States
| | - Xiangjing Meng
- Shandong Academy of Pharmaceutical Science, Jinan, China
| | - Dongmei Li
- Department of Microbiology & Immunology, Georgetown University Medical Center, Washington, DC, United States
| | - Richard Calderone
- Department of Microbiology & Immunology, Georgetown University Medical Center, Washington, DC, United States
| | - Dewei Mao
- Sport Science Research Center, Shandong Sport University, Jinan, China
| | - Bo Sui
- Sport Science Research Center, Shandong Sport University, Jinan, China
| |
Collapse
|
804
|
Lee JH, Wen X, Cho H, Koo SH. CREB/CRTC2 controls GLP-1-dependent regulation of glucose homeostasis. FASEB J 2018; 32:1566-1578. [PMID: 29118086 DOI: 10.1096/fj.201700845r] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Glucagon-like peptide 1 (GLP-1) is a major incretin that controls glucose homeostasis. The secretion of mature GLP-1 is regulated via GPCRs, including bile acid receptor G protein-coupled bile acid receptor 1, which uses cAMP signaling to enhance the exocytosis of GLP-1-containing vesicles. However, the role of cAMP-mediated transcription has not been clearly demonstrated to date. In this study, we explored the role of cAMP response element-binding protein/CREB-regulated transcription coactivator 2 (CREB/CRTC2)-dependent transcription on GLP-1 secretion in the L cells. We found that the reduced CREB/CRTC2 activity impaired the cAMP-dependent increase in GLP-1 secretion, whereas expression of constitutively active CRTC2 increased GLP-1 exocytosis from the L cells. Close investigation revealed that expression of not only proglucagon but also PC1/3, an endopeptidase for GLP-1 maturation, is transcriptionally regulated by CREB/CRTC2. Furthermore, expression of peroxisome proliferator-activating receptor coactivator 1 α is also reduced upon depletion of CRTC2, leading to the decreased expression of oxidative phosphorylation (OxPhos) genes, reduced ATP levels, and calcium concentrations in the L cells. Finally, we observed that intestine-specific CRTC2 knockout mice displayed reduced GLP-1 expression, leading to the lower plasma GLP-1 levels, impaired glucose tolerance, and decreased insulin-containing β cells in pancreatic islets. Our data show that the CREB/CRTC2-dependent transcriptional pathway is critical for regulating glucose homeostasis by controlling production of GLP-1 from the L cells at the level of transcription, maturation, and exocytosis.-Lee, J.-H., Wen, X., Cho, H., Koo, S.-H. CREB/CRTC2 controls GLP-1-dependent regulation of glucose homeostasis.
Collapse
Affiliation(s)
- Ji-Hyun Lee
- Division of Life Sciences, Korea University, Seoul, Korea
| | - Xianlan Wen
- Department of Physiology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Hana Cho
- Department of Physiology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Seung-Hoi Koo
- Division of Life Sciences, Korea University, Seoul, Korea
| |
Collapse
|
805
|
Chen Y, Wu S, Tian Y. Cholecystectomy as a risk factor of metabolic syndrome: from epidemiologic clues to biochemical mechanisms. J Transl Med 2018; 98:7-14. [PMID: 28892095 DOI: 10.1038/labinvest.2017.95] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 07/03/2017] [Accepted: 07/09/2017] [Indexed: 12/22/2022] Open
Abstract
Cholecystectomy has long been regarded as a safe procedure with no deleterious influence on the body. However, recent studies provide clues that link cholecystectomy to a high risk for metabolic syndrome (MetS). In the present review, we describe the epidemiologic evidence that links cholecystectomy to MetS. Various components of MetS are investigated, including visceral obesity, dyslipidemia, elevated blood pressure, impaired fasting glucose, and insulin resistance. The possible mechanisms that associate cholecystectomy with MetS are discussed on the basis of experimental studies.
Collapse
Affiliation(s)
- Yongsheng Chen
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shuodong Wu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu Tian
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
806
|
Wang M, Chen Y, Wang Y, Li Y, Zheng H, Ma F, Ma C, Zhang X, Lu B, Xie Z, Liao Q. The effect of probiotics and polysaccharides on the gut microbiota composition and function of weaned rats. Food Funct 2018. [DOI: 10.1039/c7fo01507k] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A combination of probiotics and polysaccharides may be used as a functional food to modulate the composition and function of gut microbiota.
Collapse
Affiliation(s)
- Mengxia Wang
- School of Pharmaceutical Sciences
- Guangzhou University of Chinese Medicine
- Guangzhou
- P. R. China
| | - Yongxiong Chen
- School of Pharmaceutical Sciences
- Guangzhou University of Chinese Medicine
- Guangzhou
- P. R. China
| | | | - Yuan Li
- School of Pharmaceutical Sciences
- Guangzhou University of Chinese Medicine
- Guangzhou
- P. R. China
| | - Haihui Zheng
- School of Pharmaceutical Sciences(Shenzhen)
- Sun Yat-sen University
- Guangzhou
- P. R. China
| | - Fangli Ma
- Infinitus (China) Company Ltd
- Guangzhou
- China
| | | | - Xiaojun Zhang
- School of Pharmaceutical Sciences
- Guangzhou University of Chinese Medicine
- Guangzhou
- P. R. China
| | - Biyu Lu
- School of Pharmaceutical Sciences
- Guangzhou University of Chinese Medicine
- Guangzhou
- P. R. China
| | - Zhiyong Xie
- School of Pharmaceutical Sciences(Shenzhen)
- Sun Yat-sen University
- Guangzhou
- P. R. China
| | - Qiongfeng Liao
- School of Pharmaceutical Sciences
- Guangzhou University of Chinese Medicine
- Guangzhou
- P. R. China
| |
Collapse
|
807
|
Ikegami T, Honda A. Reciprocal interactions between bile acids and gut microbiota in human liver diseases. Hepatol Res 2018; 48:15-27. [PMID: 29150974 DOI: 10.1111/hepr.13001] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 11/04/2017] [Accepted: 11/06/2017] [Indexed: 02/08/2023]
Abstract
The gut microbiota (GM) play a central role in their host's metabolism of bile acids (BAs) by regulating deconjugation, dehydroxylation, dehydrogenation, and epimerization reactions to generate unconjugated free BAs and secondary BAs. These BAs generated by the GM are potent signaling molecules that interact with BA receptors, such as the farnesoid X receptor and Takeda G-protein-coupled receptor 5. Each BA has a differential affinity to these receptors; therefore, alterations in BA composition by GM could modify the intensity of receptor signaling. Bile acids also act as antimicrobial agents by damaging bacterial membranes and as detergents by altering intracellular macromolecular structures. Therefore, BAs and the GM reciprocally control each other's compositions. In this review, we discuss the latest findings on the mutual effects of BAs and GM on each other; we also describe their roles in the pathophysiology of liver disease progression and potential therapeutic applications of targeting this cross-talk.
Collapse
Affiliation(s)
- Tadashi Ikegami
- Division of Gastroenterology and Hepatology, Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan
| | - Akira Honda
- Division of Gastroenterology and Hepatology, Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan.,Joint Research Center, Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan
| |
Collapse
|
808
|
Wang XX, Wang D, Luo Y, Myakala K, Dobrinskikh E, Rosenberg AZ, Levi J, Kopp JB, Field A, Hill A, Lucia S, Qiu L, Jiang T, Peng Y, Orlicky D, Garcia G, Herman-Edelstein M, D'Agati V, Henriksen K, Adorini L, Pruzanski M, Xie C, Krausz KW, Gonzalez FJ, Ranjit S, Dvornikov A, Gratton E, Levi M. FXR/TGR5 Dual Agonist Prevents Progression of Nephropathy in Diabetes and Obesity. J Am Soc Nephrol 2018; 29:118-137. [PMID: 29089371 PMCID: PMC5748904 DOI: 10.1681/asn.2017020222] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 08/23/2017] [Indexed: 12/12/2022] Open
Abstract
Bile acids are ligands for the nuclear hormone receptor farnesoid X receptor (FXR) and the G protein-coupled receptor TGR5. We have shown that FXR and TGR5 have renoprotective roles in diabetes- and obesity-related kidney disease. Here, we determined whether these effects are mediated through differential or synergistic signaling pathways. We administered the FXR/TGR5 dual agonist INT-767 to DBA/2J mice with streptozotocin-induced diabetes, db/db mice with type 2 diabetes, and C57BL/6J mice with high-fat diet-induced obesity. We also examined the individual effects of the selective FXR agonist obeticholic acid (OCA) and the TGR5 agonist INT-777 in diabetic mice. The FXR agonist OCA and the TGR5 agonist INT-777 modulated distinct renal signaling pathways involved in the pathogenesis and treatment of diabetic nephropathy. Treatment of diabetic DBA/2J and db/db mice with the dual FXR/TGR5 agonist INT-767 improved proteinuria and prevented podocyte injury, mesangial expansion, and tubulointerstitial fibrosis. INT-767 exerted coordinated effects on multiple pathways, including stimulation of a signaling cascade involving AMP-activated protein kinase, sirtuin 1, PGC-1α, sirtuin 3, estrogen-related receptor-α, and Nrf-1; inhibition of endoplasmic reticulum stress; and inhibition of enhanced renal fatty acid and cholesterol metabolism. Additionally, in mice with diet-induced obesity, INT-767 prevented mitochondrial dysfunction and oxidative stress determined by fluorescence lifetime imaging of NADH and kidney fibrosis determined by second harmonic imaging microscopy. These results identify the renal signaling pathways regulated by FXR and TGR5, which may be promising targets for the treatment of nephropathy in diabetes and obesity.
Collapse
MESH Headings
- Albuminuria/etiology
- Animals
- Bile Acids and Salts/pharmacology
- Chenodeoxycholic Acid/analogs & derivatives
- Chenodeoxycholic Acid/pharmacology
- Cholesterol/metabolism
- Cholic Acids/pharmacology
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Type 2/metabolism
- Diabetic Nephropathies/complications
- Diabetic Nephropathies/metabolism
- Diabetic Nephropathies/pathology
- Diabetic Nephropathies/prevention & control
- Disease Progression
- Endoplasmic Reticulum Stress
- Fibrosis
- Glomerular Mesangium/pathology
- Humans
- Kidney Tubules/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Mitochondria/metabolism
- Obesity/complications
- Obesity/metabolism
- Oxidative Stress
- Podocytes/pathology
- RNA, Messenger/metabolism
- Receptors, Cytoplasmic and Nuclear/agonists
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, G-Protein-Coupled/agonists
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Signal Transduction/drug effects
- Triglycerides/metabolism
Collapse
Affiliation(s)
- Xiaoxin X Wang
- Departments of Medicine and
- Pathology, University of Colorado Denver and Department of Veterans Affairs Medical Center, Aurora, Colorado
| | - Dong Wang
- Departments of Medicine and
- Pathology, University of Colorado Denver and Department of Veterans Affairs Medical Center, Aurora, Colorado
| | - Yuhuan Luo
- Departments of Medicine and
- Pathology, University of Colorado Denver and Department of Veterans Affairs Medical Center, Aurora, Colorado
| | - Komuraiah Myakala
- Departments of Medicine and
- Pathology, University of Colorado Denver and Department of Veterans Affairs Medical Center, Aurora, Colorado
| | - Evgenia Dobrinskikh
- Departments of Medicine and
- Pathology, University of Colorado Denver and Department of Veterans Affairs Medical Center, Aurora, Colorado
| | - Avi Z Rosenberg
- National Institute of Diabetes and Digestive and Kidney Diseases and
- Division of Pathology and
| | - Jonathan Levi
- National Institute of Diabetes and Digestive and Kidney Diseases and
| | - Jeffrey B Kopp
- National Institute of Diabetes and Digestive and Kidney Diseases and
| | - Amanda Field
- Center for Genetic Medicine Research, Children's Research Institute, Children's National Medical Center and the George Washington University School of Medicine and Health Sciences, Washington, DC
- Department of Nephrology and Hypertension, Rabin Medical Center, Tel Aviv, Israel
| | - Ashley Hill
- Center for Genetic Medicine Research, Children's Research Institute, Children's National Medical Center and the George Washington University School of Medicine and Health Sciences, Washington, DC
- Department of Nephrology and Hypertension, Rabin Medical Center, Tel Aviv, Israel
| | - Scott Lucia
- Departments of Medicine and
- Pathology, University of Colorado Denver and Department of Veterans Affairs Medical Center, Aurora, Colorado
| | - Liru Qiu
- Departments of Medicine and
- Pathology, University of Colorado Denver and Department of Veterans Affairs Medical Center, Aurora, Colorado
| | - Tao Jiang
- Departments of Medicine and
- Pathology, University of Colorado Denver and Department of Veterans Affairs Medical Center, Aurora, Colorado
| | - Yingqiong Peng
- Departments of Medicine and
- Pathology, University of Colorado Denver and Department of Veterans Affairs Medical Center, Aurora, Colorado
| | - David Orlicky
- Departments of Medicine and
- Pathology, University of Colorado Denver and Department of Veterans Affairs Medical Center, Aurora, Colorado
| | - Gabriel Garcia
- Departments of Medicine and
- Pathology, University of Colorado Denver and Department of Veterans Affairs Medical Center, Aurora, Colorado
| | - Michal Herman-Edelstein
- Department of Nephrology and Hypertension, Rabin Medical Center, Tel Aviv, Israel
- Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Vivette D'Agati
- Department of Pathology, Columbia University College of Physicians and Surgeons, New York, New York
| | - Kammi Henriksen
- Department of Pathology, University of Chicago, Chicago, Illinois
| | | | - Mark Pruzanski
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Cen Xie
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Kristopher W Krausz
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Frank J Gonzalez
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Suman Ranjit
- Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, California
| | - Alexander Dvornikov
- Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, California
| | - Enrico Gratton
- Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, California
| | - Moshe Levi
- Departments of Medicine and
- Pathology, University of Colorado Denver and Department of Veterans Affairs Medical Center, Aurora, Colorado
| |
Collapse
|
809
|
Aw W, Fukuda S. Understanding the role of the gut ecosystem in diabetes mellitus. J Diabetes Investig 2018; 9:5-12. [PMID: 28390093 PMCID: PMC5754518 DOI: 10.1111/jdi.12673] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Revised: 02/23/2017] [Accepted: 04/05/2017] [Indexed: 12/14/2022] Open
Abstract
Diabetes mellitus is a type of metabolic disorder whereby patients are unable to regulate glycemia. It is currently a worldwide public health issue, and is a burden to society because of its disabling and common complications. Diabetes is multifactorial, and also induces the onset of other diseases. In the present report, we review the labyrinth encompassing the gut microbiota and gut microbiota-derived metabolites in type 1 diabetes and type 2 diabetes pathogenesis. There have been exceptional improvements in deoxyribonucleic acid sequencing and mass spectrometry technologies throughout these past years, and these have allowed the comprehensive collection of information on our unique gut ecosystem. We would like to advocate incorporating metagenome and metabolome information for a comprehensive perspective of the complex interrelationships between the gut environment, host metabolism and diabetes pathogenesis. We hope that with this improved understanding we would be able to provide exciting novel therapeutic approaches to engineer an ideal gut ecosystem for optimal health.
Collapse
Affiliation(s)
- Wanping Aw
- Institute for Advanced BiosciencesKeio UniversityTsuruokaYamagataJapan
| | - Shinji Fukuda
- Institute for Advanced BiosciencesKeio UniversityTsuruokaYamagataJapan
- PRESTOJapan Science and Technology AgencySaitamaJapan
| |
Collapse
|
810
|
Molinaro A, Wahlström A, Marschall HU. Role of Bile Acids in Metabolic Control. Trends Endocrinol Metab 2018; 29:31-41. [PMID: 29195686 DOI: 10.1016/j.tem.2017.11.002] [Citation(s) in RCA: 313] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 11/04/2017] [Accepted: 11/06/2017] [Indexed: 02/07/2023]
Abstract
Bile acids are endocrine molecules that in addition to facilitating the absorption of fat-soluble nutrients regulate numerous metabolic processes, including glucose, lipid, and energy homeostasis. The signaling actions of bile acids are mediated through specific bile-acid-activated nuclear and membrane-bound receptors. These receptors are not only expressed by tissues within the enterohepatic circulation such as the liver and the intestine, but also in other organs where bile acids mediate their systemic actions. In this review, we discuss bile acid signaling and the interplay with the gut microbiota in the pathophysiology of obesity, type 2 diabetes, and non-alcoholic fatty liver disease, and the role of surgical and pharmacological interventions on bile acid profiles and metabolism.
Collapse
Affiliation(s)
- Antonio Molinaro
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, S-413 45 Gothenburg, Sweden
| | - Annika Wahlström
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, S-413 45 Gothenburg, Sweden
| | - Hanns-Ulrich Marschall
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, S-413 45 Gothenburg, Sweden.
| |
Collapse
|
811
|
Shima KR, Ota T, Kato KI, Takeshita Y, Misu H, Kaneko S, Takamura T. Ursodeoxycholic acid potentiates dipeptidyl peptidase-4 inhibitor sitagliptin by enhancing glucagon-like peptide-1 secretion in patients with type 2 diabetes and chronic liver disease: a pilot randomized controlled and add-on study. BMJ Open Diabetes Res Care 2018; 6:e000469. [PMID: 29607050 PMCID: PMC5873545 DOI: 10.1136/bmjdrc-2017-000469] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 02/13/2018] [Accepted: 02/20/2018] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE We evaluated the effects of ursodeoxycholic acid (UDCA) on glucagon-like peptide-1 (GLP-1) secretion and glucose tolerance in patients with type 2 diabetes with chronic liver disease. RESEARCH DESIGN AND METHODS Japanese patients with type 2 diabetes (glycated hemoglobin (HbA1c) levels ≥7.0%) and chronic liver disease were included in this study. Sixteen patients (HbA1c level, 7.2%±0.6%(55.2 mmol/mol)) were randomized to receive 900 mg UDCA for 12 weeks followed by 50 mg sitagliptin add-on therapy for 12 weeks (UDCA-first group; n=8) or 50 mg sitagliptin for 12 weeks followed by 900 mg UDCA add-on therapy for 12 weeks (sitagliptin-first group; n=8). All patients underwent a liquid high-fat meal test before and after 12 or 24 weeks of treatment. RESULTS The baseline characteristics were similar between the UDCA-first and sitagliptin-first groups. There was a decrease in body weight (72.5±8.4 to 70.6±8.6 kg; P=0.04) and the HbA1c level (7.0%±0.3% to 6.4%±0.5%(53.0 to 46.4 mmol/mol); P=0.01) in the UDCA-first group. The HbA1c level decreased further after sitagliptin administration (6.4%±0.5% to 6.0%±0.4%(46.4 to 42.1 mmol/mol); P<0.01). Although there were no initial changes in the weight and HbA1c level in the sitagliptin-first group, the HbA1c level decreased after UDCA addition (7.1%±1.1% to 6.6%±0.9%(54.1 to 48.6 mmol/mol); P=0.04). UDCA alone increased the area under the curve0-30 for GLP-1 response (115.4±47.2 to 221.9±48.9 pmol·min/L; P<0.01), but not the glucose-dependent insulinotropic polypeptide response, in the UDCA-first group. CONCLUSIONS UDCA treatment resulted in a greater reduction in HbA1c levels, and an increased early phase GLP-1 secretion. TRIAL REGISTRATION NUMBER NCT01337440.
Collapse
Affiliation(s)
- Kosuke Robert Shima
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Tsuguhito Ota
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Ken-ichiro Kato
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yumie Takeshita
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Hirofumi Misu
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Shuichi Kaneko
- Department of System Biology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Toshinari Takamura
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
812
|
An Overview of the Roles of the Gut Microbiome in Obesity and Diabetes. NUTRITIONAL AND THERAPEUTIC INTERVENTIONS FOR DIABETES AND METABOLIC SYNDROME 2018. [DOI: 10.1016/b978-0-12-812019-4.00006-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
813
|
Mazidi M, de Caravatto PPP, Speakman JR, Cohen RV. Mechanisms of Action of Surgical Interventions on Weight-Related Diseases: the Potential Role of Bile Acids. Obes Surg 2017; 27:826-836. [PMID: 28091894 DOI: 10.1007/s11695-017-2549-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Surgical interventions for weight-related diseases (SWRD) may have substantial and sustainable effect on weight reduction, also leading to a higher remission rate of type 2 diabetes (T2D) mellitus than any other medical treatment or lifestyle intervention. The resolution of T2D after Roux-en-Y gastric bypass (RYGB) typically occurs too quickly to be accounted for by weight loss alone, suggesting that these operations have a direct impact on glucose homeostasis. The mechanisms underlying these beneficial effects however remain unclear. Recent research suggests that changes in the concentrations of plasma bile acids might contribute to these metabolic changes after surgery. In this review, we aimed to outline the potential role of bile acids in SWRD. We systematically reviewed MEDLINE, SCOPUS, and Web of Science for articles reporting the effect of SWRD on outcomes published between 1969 and 2016. We found that changes in circulating bile acids after surgery may play a major role through activation of the farnesoid X receptor A (FXRA), the fibroblast growth factor 19 (FGF19), and the G protein-coupled bile acid receptor (TGR5). Bile acid concentration increased significantly after RYGB. Some studies suggest that a transitory decrease occurs at 1 week post-surgery, followed by a gradual increase. Most studies have shown the increase to be proportionate by all bile acid subtypes. Bile acids can regulate glucose metabolism through the expression of TGR5 receptor in L cells, resulting in a release of glucagon-like peptide 1 (GLP-1). It may also induce the synthesis and secretion of FGF19 in ileal cells, thereby improving insulin sensitivity and regulating glucose metabolism. All the present SWRD are involved with changes in food stimulation to the stomach. This implies that discovering and developing the antagonists to TGR5 and FXRA may effectively control metabolic syndrome and the elucidation of the mechanisms underlying the physiological effects related to weight loss and T2D remission after surgery may help to identify new drug targets.
Collapse
Affiliation(s)
- Mohsen Mazidi
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Chaoyang, Beijing, China.,University of the Chinese Academy of Sciences, Huairou, Beijing, China
| | - Pedro Paulo P de Caravatto
- The Center for Obesity and Diabetes, Oswaldo Cruz German Hospital, Rua Cincinato Braga, 37 5o. andar, São Paulo, São Paulo, Brazil
| | - John R Speakman
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Chaoyang, Beijing, China.,Institute of Biological and Environmental Science, University of Aberdeen, Aberdeen, Scotland, UK
| | - Ricardo V Cohen
- The Center for Obesity and Diabetes, Oswaldo Cruz German Hospital, Rua Cincinato Braga, 37 5o. andar, São Paulo, São Paulo, Brazil.
| |
Collapse
|
814
|
Akinrotimi O, Riessen R, VanDuyne P, Park JE, Lee YK, Wong LJ, Zavacki AM, Schoonjans K, Anakk S. Small heterodimer partner deletion prevents hepatic steatosis and when combined with farnesoid X receptor loss protects against type 2 diabetes in mice. Hepatology 2017; 66:1854-1865. [PMID: 28586124 PMCID: PMC5696047 DOI: 10.1002/hep.29305] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 05/05/2017] [Accepted: 06/01/2017] [Indexed: 02/06/2023]
Abstract
UNLABELLED Nuclear receptors farnesoid X receptor (FXR) and small heterodimer partner (SHP) are important regulators of bile acid, lipid, and glucose homeostasis. Here, we show that global Fxr -/- Shp-/- double knockout (DKO) mice are refractory to weight gain, glucose intolerance, and hepatic steatosis when challenged with high-fat diet. DKO mice display an inherently increased capacity to burn fat and suppress de novo hepatic lipid synthesis. Moreover, DKO mice were also very active and that correlated well with the observed increase in phosphoenolpyruvate carboxykinase expression, type IA fibers, and mitochondrial function in skeletal muscle. Mechanistically, we demonstrate that liver-specific Shp deletion protects against fatty liver development by suppressing expression of peroxisome proliferator-activated receptor gamma 2 and lipid-droplet protein fat-specific protein 27 beta. CONCLUSION These data suggest that Fxr and Shp inactivation may be beneficial to combat diet-induced obesity and uncover that hepatic SHP is necessary to promote fatty liver disease. (Hepatology 2017;66:1854-1865).
Collapse
Affiliation(s)
- Oludemilade Akinrotimi
- Department of Molecular and Integrative Physiology, University of Illinois, Urbana-Champaign, Urbana, Il 61801
| | - Ryan Riessen
- Department of Molecular and Integrative Physiology, University of Illinois, Urbana-Champaign, Urbana, Il 61801
| | - Philip VanDuyne
- Department of Molecular and Integrative Physiology, University of Illinois, Urbana-Champaign, Urbana, Il 61801
| | - Jung Eun Park
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272
| | - Yoon Kwang Lee
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272
| | - Lee-Jun Wong
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030
| | - Ann M Zavacki
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Kristina Schoonjans
- Laboratory of Metabolic Signaling, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Sayeepriyadarshini Anakk
- Department of Molecular and Integrative Physiology, University of Illinois, Urbana-Champaign, Urbana, Il 61801,To whom correspondence should be addressed
| |
Collapse
|
815
|
Herbert KE, Erridge C. Regulation of low-density lipoprotein cholesterol by intestinal inflammation and the acute phase response. Cardiovasc Res 2017; 114:226-232. [DOI: 10.1093/cvr/cvx237] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 11/29/2017] [Indexed: 02/07/2023] Open
Abstract
AbstractSystemic inflammation, induced by disease or experimental intervention, is well established to result in elevated levels of circulating triglycerides, and reduced levels of high-density lipoprotein-cholesterol (HDL-C), in most mammalian species. However, the relationship between inflammation and low-density lipoprotein-cholesterol (LDL-C) concentrations is less clear. Most reports indicate that systemic inflammation, as observed during sepsis or following high dose experimental endotoxaemia, lowers total, and LDL-C in man. However, isolated reports have suggested that certain inflammatory conditions are associated with increased LDL-C. In this review, we summarize the emerging evidence that low-grade inflammation specifically of intestinal origin may be associated with increased serum LDL-C levels. Preliminary insights into potential mechanisms that may mediate these effects, including those connecting inflammation to trans-intestinal cholesterol efflux (TICE), are considered. We conclude that this evidence supports the potential downregulation of major mediators of TICE by inflammatory mediators in vitro and during intestinal inflammation in vivo. The TICE-inflammation axis therefore merits further study in terms of its potential to regulate serum LDL-C, and as a readily druggable target for hypercholesterolaemia.
Collapse
Affiliation(s)
- Karl E Herbert
- Department of Cardiovascular Sciences, Glenfield Hospital, University of Leicester, Groby Road, Leicester, Leicestershire, LE3 9QP, UK
| | - Clett Erridge
- Department of Cardiovascular Sciences, Glenfield Hospital, University of Leicester, Groby Road, Leicester, Leicestershire, LE3 9QP, UK
- Department of Biomedical and Forensic Sciences, Anglia Ruskin University, East Road, Cambridge, Cambridgeshire, CB1 1PT, UK
| |
Collapse
|
816
|
Heiss CN, Olofsson LE. Gut Microbiota-Dependent Modulation of Energy Metabolism. J Innate Immun 2017; 10:163-171. [PMID: 29131106 DOI: 10.1159/000481519] [Citation(s) in RCA: 187] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 09/14/2017] [Indexed: 12/24/2022] Open
Abstract
The gut microbiota has emerged as an environmental factor that modulates the host's energy balance. It increases the host's ability to harvest energy from the digested food, and produces metabolites and microbial products such as short-chain fatty acids, secondary bile acids, and lipopolysaccharides. These metabolites and microbial products act as signaling molecules that modulate appetite, gut motility, energy uptake and storage, and energy expenditure. Several findings suggest that the gut microbiota can affect the development of obesity. Germ-free mice are leaner than conventionally raised mice and they are protected against diet-induced obesity. Furthermore, obese humans and rodents have an altered gut microbiota composition with less phylogeneic diversity compared to lean controls, and transplantation of the gut microbiota from obese subjects to germ-free mice can transfer the obese phenotype. Taken together, these findings indicate a role for the gut microbiota in obesity and suggest that the gut microbiota could be targeted to improve metabolic diseases like obesity. This review focuses on the role of the gut microbiota in energy balance regulation and its potential role in obesity.
Collapse
Affiliation(s)
- Christina N Heiss
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | | |
Collapse
|
817
|
Lyu M, Wang YF, Fan GW, Wang XY, Xu SY, Zhu Y. Balancing Herbal Medicine and Functional Food for Prevention and Treatment of Cardiometabolic Diseases through Modulating Gut Microbiota. Front Microbiol 2017; 8:2146. [PMID: 29167659 PMCID: PMC5682319 DOI: 10.3389/fmicb.2017.02146] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 10/19/2017] [Indexed: 12/22/2022] Open
Abstract
It has become apparent that gut microbiota is closely associated with cardiometabolic diseases (CMDs), and alteration in microbiome compositions is also linked to the host environment. Next generation sequencing (NGS) has facilitated in-depth studies on the effects of herbal medicine and functional food on gut microbiota. Both herbal medicine and functional food contain fiber, polyphenols and polysaccharides, exerting prebiotics-like activities in the prevention and treatment of CMDs. The administrations of herbal medicine and functional food lead to increased the abundance of phylum Bacteroidetes, and genus Akkermansia, Bifidobacteria, Lactobacillus, Bacteroides and Prevotella, while reducing phylum Firmicutes and Firmicutes/Bacteroidetes ratio in gut. Both herbal medicine and functional food interact with gut microbiome and alter the microbial metabolites including short-chain fatty acids (SCFAs), bile acids (BAs) and lipopolysaccharides (LPS), which are now correlated with metabolic diseases such as type 2 diabetes (T2D), obesity and non-alcoholic fatty liver disease (NAFLD). In addition, trimethylamine (TMA)-N-oxide (TMAO) is recently linked to atherosclerosis (AS) and cardiovascular disease (CVD) risks. Moreover, gut-organs axes may serve as the potential strategy for treating CMDs with the intervention of herbal medicine and functional food. In summary, a balance between herbal medicine and functional food rich in fiber, polyphenols and polysaccharides plays a vital role in modulating gut microbiota (phylum Bacteroidetes, Firmicutes and Firmicutes/Bacteroidetes ratio, and genus Akkermansia, Bifidobacteria, Lactobacillus, Bacteroides and Prevotella) through SCFAs, BAs, LPS and TMAO signaling regarding CMDs. Targeting gut-organs axes may serve as a new therapeutic strategy for CMDs by herbal medicine and functional food in the future. This review aims to summarize the balance between herbal medicine and functional food utilized for the prevention and treatment of CMDs through modulating gut microbiota.
Collapse
Affiliation(s)
- Ming Lyu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, Tianjin, China
| | - Yue-Fei Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, Tianjin, China
| | - Guan-Wei Fan
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, Tianjin, China.,Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiao-Ying Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Neuroscience Program, Neuroprotection Research Laboratory, Department of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | | | - Yan Zhu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, Tianjin, China
| |
Collapse
|
818
|
Mertens KL, Kalsbeek A, Soeters MR, Eggink HM. Bile Acid Signaling Pathways from the Enterohepatic Circulation to the Central Nervous System. Front Neurosci 2017; 11:617. [PMID: 29163019 PMCID: PMC5681992 DOI: 10.3389/fnins.2017.00617] [Citation(s) in RCA: 189] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 10/23/2017] [Indexed: 12/14/2022] Open
Abstract
Bile acids are best known as detergents involved in the digestion of lipids. In addition, new data in the last decade have shown that bile acids also function as gut hormones capable of influencing metabolic processes via receptors such as FXR (farnesoid X receptor) and TGR5 (Takeda G protein-coupled receptor 5). These effects of bile acids are not restricted to the gastrointestinal tract, but can affect different tissues throughout the organism. It is still unclear whether these effects also involve signaling of bile acids to the central nervous system (CNS). Bile acid signaling to the CNS encompasses both direct and indirect pathways. Bile acids can act directly in the brain via central FXR and TGR5 signaling. In addition, there are two indirect pathways that involve intermediate agents released upon interaction with bile acids receptors in the gut. Activation of intestinal FXR and TGR5 receptors can result in the release of fibroblast growth factor 19 (FGF19) and glucagon-like peptide 1 (GLP-1), both capable of signaling to the CNS. We conclude that when plasma bile acids levels are high all three pathways may contribute in signal transmission to the CNS. However, under normal physiological circumstances, the indirect pathway involving GLP-1 may evoke the most substantial effect in the brain.
Collapse
Affiliation(s)
- Kim L Mertens
- Master's Program in Biomedical Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Laboratory of Endocrinology, Department Clinical Chemistry, Academic Medical Centre, University of Amsterdam, Amsterdam, Netherlands.,Department of Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| | - Maarten R Soeters
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Hannah M Eggink
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Department of Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| |
Collapse
|
819
|
Garruti G, Di Ciaula A, Wang HH, Wang DQH, Portincasa P. Cross-Talk Between Bile Acids and Gastro-Intestinal and Thermogenic Hormones: Clues from Bariatric Surgery. Ann Hepatol 2017; 16:s68-s82. [PMID: 29080342 DOI: 10.5604/01.3001.0010.5499] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 09/06/2017] [Indexed: 02/05/2023]
Abstract
Obesity is rapidly increasing and has reached epidemic features worldwide. It´s linked to insulin resistance, systemic low-grade inflammation and common pathogenic pathways with a number of comorbidities (including cancer), leading to high mortality rates. Besides change of lifestyles (diet and physical exercise) and pharmacological therapy, bariatric surgery is able to rapidly improve several metabolic and morphologic features associated with excessive fat storage, and currently represents an in vivo model to study the pathogenic mechanisms underlying obesity and obesity-related complications. Studies on obese subjects undergoing bariatric surgery find that the effects of surgery are not simply secondary to gastric mechanical restriction and malabsorption which induce body weight loss. In fact, some surgical procedures positively modify key pathways involving the intestine, bile acids, receptor signaling, gut microbiota, hormones and thermogenesis, leading to systemic metabolic changes. Furthermore, bariatric surgery represents a suitable model to evaluate the gene-environment interaction and some epigenetic mechanisms linking obesity and insulin resistance to metabolic diseases.
Collapse
Affiliation(s)
- Gabriella Garruti
- Department of Emergency and Organ Transplants, Unit of Endocrinology, University of Bari Medical School, Bari, Italy
| | | | - Helen H Wang
- Department of Medicine, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - David Q-H Wang
- Department of Medicine, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Piero Portincasa
- Department of Biomedical Sciences and Human Oncology, Clinica Medica "A. Murri", University of Bari Medical School, Bari, Italy
| |
Collapse
|
820
|
Abstract
IN BRIEF Bariatric surgery is the most efficacious treatment for obesity, type 2 diabetes, and other obesity-related comorbidities. In this article, the authors review the current indications for bariatric surgery and discuss the most commonly performed procedures. They analyze medical outcomes of bariatric procedures by reviewing key prospective trials and discuss changes in physiology after these procedures. They conclude by discussing long-term management of bariatric patients by reviewing current guidelines for nutritional support and listing common complications related to these procedures.
Collapse
Affiliation(s)
- Scott Kizy
- Department of Surgery, University of Minnesota, Minneapolis, MN
| | - Cyrus Jahansouz
- Department of Surgery, University of Minnesota, Minneapolis, MN
| | - Keith Wirth
- Department of Surgery, University of Minnesota, Minneapolis, MN
| | | | - Daniel Leslie
- Department of Surgery, University of Minnesota, Minneapolis, MN
| |
Collapse
|
821
|
Lo SH, Li Y, Cheng KC, Niu CS, Cheng JT, Niu HS. Ursolic acid activates the TGR5 receptor to enhance GLP-1 secretion in type 1-like diabetic rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2017; 390:1097-1104. [PMID: 28756460 DOI: 10.1007/s00210-017-1409-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 07/21/2017] [Indexed: 12/14/2022]
Abstract
Endogenous Takeda G-protein-coupled receptor 5 (TGR5), G-protein-coupled bile acid receptor 1 (GPBAR1), regulates glucose metabolism. In animals, TGR5 activation by a chemical agonist may increase incretin secretion and reduce the blood sugar level. Recently, betulinic acid has been suggested to activate TGR5. Ursolic acid is a well-known pentacyclic triterpenoid that is similar to betulinic acid. It is of special interest to determine the potential effect of ursolic acid on TGR5. Therefore, we transfected cultured Chinese hamster ovary (CHO-K1) cells with the TGR5 gene. The functions of the transfected cells were confirmed via glucose uptake using a fluorescent indicator. Moreover, NCI-H716 cells that secreted incretin were also investigated, and the glucagon-like peptide (GLP-1) levels were quantified using ELISA kits. In addition, streptozotocin (STZ)-induced type 1-like diabetic rats were used to identify the effect of ursolic acid in vivo. Ursolic acid concentration dependently increased glucose uptake in CHO-K1 cells expressing TGR5. In NCI-H716 cells, ursolic acid induced a concentration-dependent elevation in GLP-1 secretion, which was inhibited by triamterene at the effective concentrations to block TGR5. Ursolic acid also increased the plasma GLP-1 level via TGR5 activation, which was further characterized in vivo with type 1-like diabetic rats. Moreover, ursolic acid is more effective than betulinic acid in reduction of hyperglycemia and increase of GLP-1 secretion. Therefore, we demonstrated that ursolic acid can activate TGR5, enhancing GLP-1 secretion in vitro and in vivo. Therefore, ursolic acid is suitable for use in TGR5 activation.
Collapse
Affiliation(s)
- Shih-Hsiang Lo
- Division of Cardiology, Department of Internal Medicine, Chung Hsing Branch of Taipei City Hospital, Taipei City, Taiwan, 10341
- Department of Nursing, Tzu Chi University of Science and Technology, Hualien City, Taiwan, 97005
| | - Yingxiao Li
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, 890-8520, Japan
- Department of Medical Research, Chi-Mei Medical Center, Yong Kang, Tainan City, Taiwan, 71003
| | - Kai Chun Cheng
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, 890-8520, Japan
| | - Chiang-Shan Niu
- Department of Nursing, Tzu Chi University of Science and Technology, Hualien City, Taiwan, 97005
| | - Juei-Tang Cheng
- Department of Medical Research, Chi-Mei Medical Center, Yong Kang, Tainan City, Taiwan, 71003.
- College of Health Science, Chang Jung Christian University, Institute of Medical Science, Guei-Ren, Tainan City, Taiwan, 71101.
| | - Ho-Shan Niu
- Department of Nursing, Tzu Chi University of Science and Technology, Hualien City, Taiwan, 97005
| |
Collapse
|
822
|
Brønden A, Albér A, Rohde U, Rehfeld JF, Holst JJ, Vilsbøll T, Knop FK. Single-Dose Metformin Enhances Bile Acid-Induced Glucagon-Like Peptide-1 Secretion in Patients With Type 2 Diabetes. J Clin Endocrinol Metab 2017; 102:4153-4162. [PMID: 28938439 DOI: 10.1210/jc.2017-01091] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 08/15/2017] [Indexed: 12/31/2022]
Abstract
CONTEXT Despite a position as the first-line pharmacotherapy in type 2 diabetes, the glucose-lowering mechanisms of metformin remain to be fully clarified. Gut-derived modes of action, including suppression of bile acid reabsorption and a resulting increase in glucagon-like peptide-1 (GLP-1) secretion, have been proposed. OBJECTIVE The aim of this study was to assess the GLP-1 secretory and glucometabolic effects of endogenously released bile, with and without concomitant single-dose administration of metformin in patients with type 2 diabetes. DESIGN Randomized, placebo-controlled, and double-blinded crossover study. SETTING This study was conducted at Center for Diabetes Research, Gentofte Hospital, Denmark. PATIENTS Fifteen metformin-treated patients with type 2 diabetes; all participants completed the study. INTERVENTIONS Four experimental study days in randomized order with administration of either 1500 mg metformin or placebo in combination with intravenous infusion of cholecystokinin (0.4 pmol × kg-1 × min-1) or saline. MAIN OUTCOME MEASURE Plasma GLP-1 excursions as measured by baseline-subtracted area under the curve. RESULTS Single-dose metformin further enhanced bile acid-mediated induction of GLP-1 secretion (P = 0.02), whereas metformin alone did not increase plasma GLP-1 concentrations compared with placebo (P = 0.17). Metformin, both with (P = 0.02) and without (P = 0.02) concomitant cholecystokinin-induced gallbladder emptying, elicited reduced plasma glucose excursions compared with placebo. No GLP-1-mediated induction of insulin secretion or suppression of glucagon was observed. CONCLUSIONS Metformin elicited an enhancement of the GLP-1 response to cholecystokinin-induced gallbladder emptying in patients with type 2 diabetes, whereas no derived effects on insulin or glucagon secretion were evident in this acute setting.
Collapse
Affiliation(s)
- Andreas Brønden
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, 2900 Hellerup, Denmark
| | - Anders Albér
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, 2900 Hellerup, Denmark
| | - Ulrich Rohde
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, 2900 Hellerup, Denmark
| | - Jens F Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, 2100 Copenhagen Ø, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Tina Vilsbøll
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, 2900 Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
- Steno Diabetes Center Copenhagen, University of Copenhagen, 2820 Gentofte, Denmark
| | - Filip K Knop
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, 2900 Hellerup, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| |
Collapse
|
823
|
Peng Z, Zhang Q, Mao Z, Wang J, Liu C, Lin X, Li X, Ji W, Fan J, Wang M, Su C. A rapid quantitative analysis of bile acids, lysophosphatidylcholines and polyunsaturated fatty acids in biofluids based on ultraperformance liquid chromatography coupled with triple quadrupole tandem massspectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2017; 1068-1069:343-351. [DOI: 10.1016/j.jchromb.2017.10.066] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 10/24/2017] [Accepted: 10/30/2017] [Indexed: 12/24/2022]
|
824
|
Ramírez-Pérez O, Cruz-Ramón V, Chinchilla-López P, Méndez-Sánchez N. The Role of the Gut Microbiota in Bile Acid Metabolism. Ann Hepatol 2017; 16:s15-s20. [PMID: 29080339 DOI: 10.5604/01.3001.0010.5494] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 09/12/2017] [Indexed: 02/04/2023]
Abstract
The gut microbiota has been considered a cornerstone of maintaining the health status of its human host because it not only facilitates harvesting of nutrients and energy from ingested food, but also produces numerous metabolites that can regulate host metabolism. One such class of metabolites, the bile acids, are synthesized from cholesterol in the liver and further metabolized by the gut microbiota into secondary bile acids. These bioconversions modulate the signaling properties of bile acids through the nuclear farnesoid X receptor and the G protein-coupled membrane receptor 5, which regulate diverse metabolic pathways in the host. In addition, bile acids can regulate gut microbial composition both directly and indirectly by activation of innate immune response genes in the small intestine. Therefore, host metabolism can be affected by both microbial modifications of bile acids, which leads to altered signaling via bile acid receptors, and by alterations in the composition of the microbiota. In this review, we mainly describe the interactions between bile acids and intestinal microbiota and their roles in regulating host metabolism, but we also examine the impact of bile acid composition in the gut on the intestinal microbiome and on host physiology.
Collapse
Affiliation(s)
| | - Vania Cruz-Ramón
- Liver Research Unit, Medica Sur Clinic & Foundation, Mexico City, Mexico
| | | | | |
Collapse
|
825
|
Wang LY, Cheng KC, Li Y, Niu CS, Cheng JT, Niu HS. Glycyrrhizic acid increases glucagon like peptide-1 secretion via TGR5 activation in type 1-like diabetic rats. Biomed Pharmacother 2017; 95:599-604. [PMID: 28881290 DOI: 10.1016/j.biopha.2017.08.087] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 08/13/2017] [Accepted: 08/23/2017] [Indexed: 12/25/2022] Open
Abstract
Glycyrrhizic acid (GA) is belonged to triterpenoid saponin that is contained in the root of licorice and is known to affect metabolic regulation. Recently, glucagon like peptide-1 (GLP-1) has widely been applied in diabetes therapeutics. However, the role of GLP-1 in GA-induced anti-diabetic effects is still unknown. Therefore, we are interested in understanding the association of GLP-1 with GA-induced effects. In type 1-like diabetic rats induced by streptozotocin (STZ-treated rats), GA increased the level of plasma GLP-1, which was blocked by triamterene at a dose sufficient to inhibit Takeda G-protein-coupled receptor 5 (TGR5). The direct effect of GA on TGR5 has been identified using the cultured Chinese hamster ovary cells (CHO-K1 cells) transfected TGR5 gene. Moreover, in intestinal NCI-H716 cells that secreted GLP-1, GA promoted GLP-1 secretion with a marked elevation of calcium levels. However, both effects of GA were reduced by ablation of TGR5 with siRNA in NCI-H716 cells. Therefore, we demonstrated that GA can enhance GLP-1 secretion through TGR5 activation.
Collapse
Affiliation(s)
- Lin-Yu Wang
- Department of Childhood Education and Nursery, Chia Nan University of Pharmacy and Science, Rende, Tainan City 71710, Taiwan; Division of Pediatrics, Chi-Mei Medical Center, Yong Kang, Tainan City 71003, Taiwan; Department of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung City 81201, Taiwan
| | - Kai Chun Cheng
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8520, Japan
| | - Yingxiao Li
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8520, Japan; Department of Medical Research, Chi-Mei Medical Center, Yong Kang, Tainan City 71003, Taiwan
| | - Chiang-Shan Niu
- Department of Nursing, Tzu Chi University of Science and Technology, Hualien City 97005, Taiwan
| | - Juei-Tang Cheng
- Department of Medical Research, Chi-Mei Medical Center, Yong Kang, Tainan City 71003, Taiwan; Institute of Medical Science, College of Health Science, Chang Jung Christian University, Guei-Ren, Tainan City 71101, Taiwan.
| | - Ho-Shan Niu
- Department of Nursing, Tzu Chi University of Science and Technology, Hualien City 97005, Taiwan.
| |
Collapse
|
826
|
Wang LY, Cheng KC, Li Y, Niu CS, Cheng JT, Niu HS. The Dietary Furocoumarin Imperatorin Increases Plasma GLP-1 Levels in Type 1-Like Diabetic Rats. Nutrients 2017; 9:1192. [PMID: 29084156 PMCID: PMC5707664 DOI: 10.3390/nu9111192] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 10/26/2017] [Accepted: 10/26/2017] [Indexed: 12/22/2022] Open
Abstract
Imperatorin, a dietary furocoumarin, is found not only in medicinal plants, but also in popular culinary herbs, such as parsley and fennel. Recently, imperatorin has been shown to activate GPR119 in cells. Another GPR, GPR131, also called TGR5 or G-protein-coupled bile acid receptor 1 (GPBAR1), is known to regulate glucose metabolism. Additionally, TGR5 activation increases glucagon-like peptide (GLP-1) secretion to lower blood sugar levels in animals. Therefore, the present study aims to determine whether the effects of imperatorin on GLP-1 secretion are mediated by TGR5. First, we transfected cultured Chinese hamster ovary cells (CHO-K1 cells) with the TGR5 gene. Glucose uptake was confirmed in the transfected cells using a fluorescent indicator. Moreover, NCI-H716 cells, which secrete GLP-1, were used to investigate the changes in calcium concentrations and GLP-1 levels. In addition, streptozotocin (STZ)-induced type 1-like diabetic rats were used to identify the effects of imperatorin in vivo. Imperatorin dose-dependently increased glucose uptake in CHO-K1 cells expressing TGR5. In STZ diabetic rats, similar to the results in NCI-H716 cells, imperatorin induced a marked increase of GLP-1 secretion that was reduced, but not totally abolished, by a dose of triamterene that inhibited TGR5. Moreover, increases in GLP-1 secretion induced by imperatorin and GPR119 activation were shown in NCI-H716 cells. We demonstrated that imperatorin induced GLP-1 secretion via activating TGR5 and GPR119. Therefore, imperatorin shall be considered as a TGR5 and GPR119 agonist.
Collapse
Affiliation(s)
- Lin-Yu Wang
- Department of Childhood Education and Nursery, Chia Nan University of Pharmacy and Science, Rende, Tainan City 71710, Taiwan.
- Division of Pediatrics, Chi-Mei Medical Center, Yong Kang, Tainan City 71003, Taiwan.
- Department of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung City 81201, Taiwan.
| | - Kai-Chun Cheng
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8520, Japan.
| | - Yingxiao Li
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8520, Japan.
- Department of Medical Research, Chi-Mei Medical Center, Yong Kang, Tainan City 71003, Taiwan.
| | - Chiang-Shan Niu
- Department of Nursing, Tzu Chi University of Science and Technology, Hualien City 97005, Taiwan.
| | - Juei-Tang Cheng
- Department of Medical Research, Chi-Mei Medical Center, Yong Kang, Tainan City 71003, Taiwan.
- Institute of Medical Science, College of Health Science, Chang Jung Christian University, Guei-Ren, Tainan City 71101, Taiwan.
| | - Ho-Shan Niu
- Department of Nursing, Tzu Chi University of Science and Technology, Hualien City 97005, Taiwan.
| |
Collapse
|
827
|
Karney A. [Microbiota and obesity]. DEVELOPMENTAL PERIOD MEDICINE 2017; 21. [PMID: 29077559 PMCID: PMC8522942 DOI: 10.34763/devperiodmed.20172103.203207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Overweight and obesity can have serious consequences that are a major public health problem, such as e.g. type 2 diabetes and cardiovascular disease. According to recent reports, gut microbiota may play an important role in the "epidemic" of obesity. The fact that intestinal microbiota may influence body weight, insulin sensitivity or glucose and lipid metabolism has led to the hypothesis that these changes may contribute to the pathogenesis of obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Alicja Karney
- Oddział Hospitalizacji Jednego Dnia, Instytut Matki i Dziecka, Warszawa, Polska,Alicja Karney Oddział Hospitalizacji Jednego Dnia, Instytut Matki i Dziecka ul. Kasprzaka 17a, 01-211 Warszawa, Polska tel. (+48-22) 32-77-104
| |
Collapse
|
828
|
Liu J, Li Y, Yang P, Wan J, Chang Q, Wang TTY, Lu W, Zhang Y, Wang Q, Yu LL. Gypenosides Reduced the Risk of Overweight and Insulin Resistance in C57BL/6J Mice through Modulating Adipose Thermogenesis and Gut Microbiota. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:9237-9246. [PMID: 28975783 DOI: 10.1021/acs.jafc.7b03382] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
This study investigated whether and how gypenosides from jiaogulan tea at 100 and 300 mg/kg/day levels could reduce the development of overweight and insulin resistance in C57 BL/6J mice fed a high-fat diet in 12 weeks. The 300 mg/kg/day gypenosides supplement significantly reduced final body weight, plasma total cholesterol, and homeostasis model assessment-estimated insulin resistance (HOMA-IR) index by 19.9%, 40%, and 36%, respectively, compared with the high-fat diet control group. Gypenosides also increased brown adipocyte tissue activity and white adipose tissue browning. The expression of genes involved in mitochondrial activity and fatty acid β-oxidation were also increased in both brown and white adipocyte tissues. In addition, gypenosides at 100 and 300 mg/kg/day levels decreased the ratio of Firmicutes to Bacteroidetes by 20% and 58.6%, respectively, and increased Akkermansia muciniphila abundance in the gut microbiota.
Collapse
Affiliation(s)
- Jie Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology & Business University (BTBU) , Beijing 100048, China
- Institute of Food and Nutraceutical Science, School of Agriculture and Biology, Shanghai Jiao Tong University , Shanghai 200240, China
| | - Yanfang Li
- Institute of Food and Nutraceutical Science, School of Agriculture and Biology, Shanghai Jiao Tong University , Shanghai 200240, China
| | - Puyu Yang
- Institute of Food and Nutraceutical Science, School of Agriculture and Biology, Shanghai Jiao Tong University , Shanghai 200240, China
| | - Jianchun Wan
- Institute of Food and Nutraceutical Science, School of Agriculture and Biology, Shanghai Jiao Tong University , Shanghai 200240, China
| | - Qimeng Chang
- Department of Surgery, Fudan University Minhang Hospital , Shanghai 201199, China
| | - Thomas T Y Wang
- Diet, Genomics, and Immunology Laboratory, Agricultural Research Service (ARS), USDA , Beltsville, Maryland 20705, United States
| | - Weiying Lu
- Institute of Food and Nutraceutical Science, School of Agriculture and Biology, Shanghai Jiao Tong University , Shanghai 200240, China
| | - Yaqiong Zhang
- Institute of Food and Nutraceutical Science, School of Agriculture and Biology, Shanghai Jiao Tong University , Shanghai 200240, China
| | - Qin Wang
- Department of Nutrition and Food Science, University of Maryland , College Park, Maryland 20742, United States
| | - Liangli Lucy Yu
- Department of Nutrition and Food Science, University of Maryland , College Park, Maryland 20742, United States
| |
Collapse
|
829
|
Comeglio P, Morelli A, Adorini L, Maggi M, Vignozzi L. Beneficial effects of bile acid receptor agonists in pulmonary disease models. Expert Opin Investig Drugs 2017; 26:1215-1228. [PMID: 28949776 DOI: 10.1080/13543784.2017.1385760] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Bile acids act as steroid hormones, controlling lipid, glucose and energy metabolism, as well as inflammation and fibrosis. Their actions are implemented through activation of nuclear (FXR, VDR, PXR) and membrane G protein-coupled (TGR5, S1PR2) receptors. Areas covered: This review discusses the potential of FXR and TGR5 as therapeutic targets in the treatment of pulmonary disorders linked to metabolism and/or inflammation. Obeticholic acid (OCA) is the most clinically advanced bile acid-derived agonist for FXR-mediated anti-inflammatory and anti-fibrotic effects. It therefore represents an attractive pharmacological approach for the treatment of lung conditions characterized by vascular and endothelial dysfunctions. Expert opinion: Inflammation, vascular remodeling and fibrotic processes characterize the progression of pulmonary arterial hypertension (PAH) and idiopathic pulmonary fibrosis (IPF). These processes are only partially targeted by the available therapeutic options and still represent a relevant medical need. The results hereby summarized demonstrate OCA efficacy in preventing experimental lung disorders, i.e. monocrotaline-induced PAH and bleomycin-induced fibrosis, by abating proinflammatory and vascular remodeling progression. TGR5 is also expressed in the lung, and targeting the TGR5 pathway, using the TGR5 agonist INT-777 or the dual FXR/TGR5 agonist INT-767, could also contribute to the treatment of pulmonary disorders mediated by inflammation and fibrosis.
Collapse
Affiliation(s)
- Paolo Comeglio
- a Department of Biomedical, Experimental and Clinical Sciences , University of Florence , Florence , Italy
| | - Annamaria Morelli
- b Department of Experimental and Clinical Medicine , University of Florence , Florence , Italy
| | | | - Mario Maggi
- a Department of Biomedical, Experimental and Clinical Sciences , University of Florence , Florence , Italy
| | - Linda Vignozzi
- a Department of Biomedical, Experimental and Clinical Sciences , University of Florence , Florence , Italy
| |
Collapse
|
830
|
Yan X, Li P, Tang Z, Feng B. The relationship between bile acid concentration, glucagon-like-peptide 1, fibroblast growth factor 15 and bile acid receptors in rats during progression of glucose intolerance. BMC Endocr Disord 2017; 17:60. [PMID: 28946907 PMCID: PMC5613331 DOI: 10.1186/s12902-017-0211-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 09/19/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Recent studies show that bile acids are involved in glucose and energy homeostasis through activation of G protein coupled membrane receptor (TGR5) and farnesoid X receptor (FXR). A few researches have explored changes of TGR5 and FXR in animals with impaired glucose regulation. This study aimed to observe changes of plasma total bile acids (TBA), glucagon-like-peptide 1 (GLP-1), fibroblast growth factor 15 (FGF15), intestinal expressions of TGR5 and FXR, and correlations between them in rats with glucose intolerance. METHODS Besides plasma fasting glucose, lipid, TBAs, alanine transaminase (ALT), active GLP-1(GLP-1A) and FGF15, a postprandial meal test was used to compare responses in glucose, insulin and GLP-1A among groups. The expressions of TGR5 and FXR in distal ileum and ascending colon were quantified by real-time PCR and western blot. RESULTS TGR5 expression was significantly decreased in distal ileum in DM group compared to other groups, and TGR5 and FXR expressions in ascending colon were also decreased in DM group compared to other groups. Correlation analysis showed correlations between TBA and GLP-1A or FGF15. GLP-1A was correlated with TGR5 mRNA expression in colon, and FGF15 was correlated with FXR mRNA expression in colon. CONCLUSIONS These results indicates that bile acid-TGR5/FXR axis contributes to glucose homeostasis.
Collapse
Affiliation(s)
- Xinfeng Yan
- Department of Endocrinology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120 China
| | - Peicheng Li
- Department of Endocrinology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120 China
| | - Zhaosheng Tang
- Department of Endocrinology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120 China
| | - Bo Feng
- Department of Endocrinology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120 China
| |
Collapse
|
831
|
Pharmacokinetics and Preliminary Pharmacodynamics of Single- and Multiple-dose Lyophilized Recombinant Glucagon-like Peptide-1 Receptor Agonist (rE-4) in Chinese Patients with Type 2 Diabetes Mellitus. Clin Drug Investig 2017; 37:1107-1115. [DOI: 10.1007/s40261-017-0569-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
832
|
de O Silva V, Lobato RV, Andrade EF, Orlando DR, Borges BDB, Zangeronimo MG, de Sousa RV, Pereira LJ. Effects of β-Glucans Ingestion on Alveolar Bone Loss, Intestinal Morphology, Systemic Inflammatory Profile, and Pancreatic β-Cell Function in Rats with Periodontitis and Diabetes. Nutrients 2017; 9:nu9091016. [PMID: 28906456 PMCID: PMC5622776 DOI: 10.3390/nu9091016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 08/31/2017] [Accepted: 09/05/2017] [Indexed: 12/19/2022] Open
Abstract
This study aimed to evaluate the effects of β-glucan ingestion (Saccharomyces cerevisiae) on the plasmatic levels of tumor necrosis factor-α (TNF-α) and interleukin-10 (IL-10), alveolar bone loss, and pancreatic β-cell function (HOMA-BF) in diabetic rats with periodontal disease (PD). Besides, intestinal morphology was determined by the villus/crypt ratio. A total of 48 Wistar rats weighing 203 ± 18 g were used. Diabetes was induced by the intraperitoneal injection of streptozotocin (80 mg/kg) and periodontal inflammation, by ligature. The design was completely randomized in a factorial scheme 2 × 2 × 2 (diabetic or not, with or without periodontitis, and ingesting β-glucan or not). The animals received β-glucan by gavage for 28 days. Alveolar bone loss was determined by scanning electron microscopy (distance between the cementoenamel junction and alveolar bone crest) and histometric analysis (bone area between tooth roots). β-glucan reduced plasmatic levels of TNF-α in diabetic animals with PD and of IL-10 in animals with PD (p < 0.05). β-glucan reduced bone loss in animals with PD (p < 0.05). In diabetic animals, β-glucan improved β-cell function (p < 0.05). Diabetic animals had a higher villus/crypt ratio (p < 0.05). In conclusion, β-glucan ingestion reduced the systemic inflammatory profile, prevented alveolar bone loss, and improved β-cell function in diabetic animals with PD.
Collapse
Affiliation(s)
- Viviam de O Silva
- Department of Veterinary Medicine, Federal University of Lavras (UFLA), Lavras 37200-000, Minas Gerais, Brazil.
| | - Raquel V Lobato
- Department of Veterinary Medicine, Federal University of Lavras (UFLA), Lavras 37200-000, Minas Gerais, Brazil.
| | - Eric F Andrade
- Department of Veterinary Medicine, Federal University of Lavras (UFLA), Lavras 37200-000, Minas Gerais, Brazil.
| | - Débora R Orlando
- Department of Veterinary Medicine, Federal University of Lavras (UFLA), Lavras 37200-000, Minas Gerais, Brazil.
| | - Bruno D B Borges
- Department of Health Sciences, Federal University of Lavras (UFLA), Lavras 37200-000, Minas Gerais, Brazil.
| | - Márcio G Zangeronimo
- Department of Veterinary Medicine, Federal University of Lavras (UFLA), Lavras 37200-000, Minas Gerais, Brazil.
| | - Raimundo V de Sousa
- Department of Veterinary Medicine, Federal University of Lavras (UFLA), Lavras 37200-000, Minas Gerais, Brazil.
| | - Luciano J Pereira
- Department of Health Sciences, Federal University of Lavras (UFLA), Lavras 37200-000, Minas Gerais, Brazil.
| |
Collapse
|
833
|
Yang IF, Jayaprakasha GK, Patil BS. In Vitro Bile Acid Binding Capacities of Red Leaf Lettuce and Cruciferous Vegetables. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:8054-8062. [PMID: 28812344 DOI: 10.1021/acs.jafc.7b02540] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
In the present study, we tested the bile acid binding capacity of red leaf lettuce, red cabbage, red kale, green kale, and Brussels sprouts through in vitro digestion process by simulating mouth, gastric, and intestinal digestion using six bile acids at physiological pH. Green and red kale exhibited significantly higher (86.5 ± 2.9 and 89.7 ± 0.9%, respectively) bile acid binding capacity compared to the other samples. Further, three different compositions of bile acids were tested to understand the effect on different health conditions. To predict the optimal dose for bile acid binding, we established a logistic relationship between kale dose and bile acid binding capacity. The results indicated that kale showed significantly higher bile acid binding capacity (82.5 ± 2.9% equivalent to 72.06 mg) at 1.5 g sample and remained constant up to 2.5 g. In addition, minimally processed (microwaved 3 min or steamed 8 min) green kale showed significantly enhanced bile acid binding capacity (91.1 ± 0.3 and 90.2 ± 0.7%, respectively) compared to lyophilized kale (85.5 ± 0.24%). Among the six bile acids tested, kale preferentially bound hydrophobic bile acids chenodeoxycholic acid and deoxycholic acid. Therefore, regular consumption of kale, especially minimally processed kale, can help excrete more bile acids and, thus, may lower the risk of hypercholesterolemia.
Collapse
Affiliation(s)
- Isabelle F Yang
- Vegetable and Fruit Improvement Center, Department of Horticultural Sciences, Texas A&M University , 1500 Research Parkway, Suite A120, College Station, Texas 77843, United States
| | - Guddadarangavvanahally K Jayaprakasha
- Vegetable and Fruit Improvement Center, Department of Horticultural Sciences, Texas A&M University , 1500 Research Parkway, Suite A120, College Station, Texas 77843, United States
| | - Bhimanagouda S Patil
- Vegetable and Fruit Improvement Center, Department of Horticultural Sciences, Texas A&M University , 1500 Research Parkway, Suite A120, College Station, Texas 77843, United States
| |
Collapse
|
834
|
Abstract
Purpose
This paper aims to summarize the available literatures, specifically in the following areas: metabolic and other side effects of aspartame; microbiota changes/dysbiosis and its effect on the gut-brain axis; changes on gut microbiota as a result of aspartame usage; metabolic effects (weight gain and glucose intolerance) of aspartame due to gut dysbiosis; and postulated effects of dysregulated microbiota-gut-brain axis on other aspartame side-effects (neurophysiological symptoms and immune dysfunction).
Design/methodology/approach
Aspartame is rapidly becoming a public health concern because of its purported side-effects especially neurophysiological symptom and immune dysregulation. It is also paradoxical that metabolic consequences including weight gain and impaired blood glucose levels have been observed in consumers. Exact mechanisms of above side-effects are unclear, and data are scarce but aspartame, and its metabolites may have caused disturbance in the microbiota-gut-brain axis.
Findings
Additional studies investigating the impact of aspartame on gut microbiota and metabolic health are needed.
Originality/value
Exact mechanism by which aspartame-induced gut dysbiosis and metabolic dysfunction requires further investigation.
Collapse
|
835
|
Kårhus ML, Brønden A, Sonne DP, Vilsbøll T, Knop FK. Evidence connecting old, new and neglected glucose-lowering drugs to bile acid-induced GLP-1 secretion: A review. Diabetes Obes Metab 2017; 19:1214-1222. [PMID: 28304141 DOI: 10.1111/dom.12946] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 03/13/2017] [Accepted: 03/14/2017] [Indexed: 12/25/2022]
Abstract
Bile acids are amphipathic water-soluble steroid-based molecules best known for their important lipid-solubilizing role in the assimilation of fat. Recently, bile acids have emerged as metabolic integrators with glucose-lowering potential. Among a variety of gluco-metabolic effects, bile acids have been demonstrated to modulate the secretion of the gut-derived incretin hormone glucagon-like peptide-1 (GLP-1), possibly via the transmembrane receptor Takeda G-protein-coupled receptor 5 and the nuclear farnesoid X receptor, in intestinal L cells. The present article critically reviews current evidence connecting established glucose-lowering drugs to bile acid-induced GLP-1 secretion, and discusses whether bile acid-induced GLP-1 secretion may constitute a new basis for understanding how metformin, inhibitors of the apical sodium-dependent bile acids transporter, and bile acid sequestrants - old, new and neglected glucose-lowering drugs - improve glucose metabolism.
Collapse
Affiliation(s)
- Martin L Kårhus
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Andreas Brønden
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - David P Sonne
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Department of Clinical Pharmacology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Tina Vilsbøll
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Faculty of Health and Medical Sciences, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center, Copenhagen, University of Copenhagen, Gentofte, Denmark
| | - Fillip K Knop
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Faculty of Health and Medical Sciences, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, The Novo Nordisk Foundation Centre for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
836
|
Li X, Shimizu Y, Kimura I. Gut microbial metabolite short-chain fatty acids and obesity. BIOSCIENCE OF MICROBIOTA FOOD AND HEALTH 2017; 36:135-140. [PMID: 29038768 PMCID: PMC5633527 DOI: 10.12938/bmfh.17-010] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 07/23/2017] [Indexed: 12/25/2022]
Abstract
Over the past decade, the gut microbiota has emerged as an essential mediator in the pathophysiology of obesity and related metabolic disorders. In this context, the reciprocal interactions of the gut microbiota structure and their metabolite profiles with host metabolism predisposing to a range of pathological conditions (e.g., insulin resistance) related to energy homeostasis have been increasingly discussed in various animal models and human cohorts. Remarkably, as the role of gut microbial metabolites as critical signaling molecules that function through the complementary host receptors has come to be appreciated, tremendous attention has been focused on the proposed diet-gut microbiota-host homeostasis axis, entailing extensive cross-disciplinary efforts in medical, pharmaceutical, and agricultural sciences. This review will discuss the recent advances in understanding the mechanisms whereby the gut microbiota modulates the effects of diet and shapes the host metabolism either towards or away from obesity and related metabolic conditions. In particular, the interactions of short chain fatty acids (SCFAs), a subset of key gut microbial metabolites, with their specific receptors will be reviewed in relation to host energy homeostatic regulation and evaluated for potential as novel therapeutic targets for diet-induced obesity.
Collapse
Affiliation(s)
- Xuan Li
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo 183-8509, Japan
| | - Yuuki Shimizu
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo 183-8509, Japan
| | - Ikuo Kimura
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo 183-8509, Japan
| |
Collapse
|
837
|
Torres-Fuentes C, Schellekens H, Dinan TG, Cryan JF. The microbiota-gut-brain axis in obesity. Lancet Gastroenterol Hepatol 2017; 2:747-756. [PMID: 28844808 DOI: 10.1016/s2468-1253(17)30147-4] [Citation(s) in RCA: 414] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 05/04/2017] [Accepted: 05/07/2017] [Indexed: 12/16/2022]
Abstract
Changes in microbial diversity and composition are increasingly associated with several disease states including obesity and behavioural disorders. Obesity-associated microbiota alter host energy harvesting, insulin resistance, inflammation, and fat deposition. Additionally, intestinal microbiota can regulate metabolism, adiposity, homoeostasis, and energy balance as well as central appetite and food reward signalling, which together have crucial roles in obesity. Moreover, some strains of bacteria and their metabolites might target the brain directly via vagal stimulation or indirectly through immune-neuroendocrine mechanisms. Therefore, the gut microbiota is becoming a target for new anti-obesity therapies. Further investigations are needed to elucidate the intricate gut-microbiota-host relationship and the potential of gut-microbiota-targeted strategies, such as dietary interventions and faecal microbiota transplantation, as promising metabolic therapies that help patients to maintain a healthy weight throughout life.
Collapse
Affiliation(s)
| | - Harriët Schellekens
- APC Microbiome Institute, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Institute, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Sciences, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Institute, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| |
Collapse
|
838
|
Ekstrand B, Young JF, Rasmussen MK. Taste receptors in the gut - A new target for health promoting properties in diet. Food Res Int 2017; 100:1-8. [PMID: 28888429 DOI: 10.1016/j.foodres.2017.08.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 08/07/2017] [Accepted: 08/12/2017] [Indexed: 12/17/2022]
Abstract
In this review we describe a new target for food functionality, the taste receptors in the gastrointestinal tract. These receptors are involved in an intricate signalling network for monitoring of taste and nutrient intake, homeostasis and energy metabolism, and they are also an early warning system for toxic substances in our diet. Especially the receptors for bitter taste provide a new possibility to activate a number of health related signalling pathways, already at low concentrations of the active substance, without requiring uptake into the body and transport via the circulation. When ligands bind to these receptors, signalling is induced either via peptide hormones into the circulation to other organs in the body, or via nerve fibers directly to the brain.
Collapse
Affiliation(s)
- Bo Ekstrand
- Chalmers University of Technology, Department of Biology and Biological Engineering, Food and Nutrition Science, SE-412 96 Gothenburg, Sweden
| | | | | |
Collapse
|
839
|
Abstract
非酒精性脂肪肝病(non-alcoholic fatty liver disease, NAFLD)是一种除饮酒以及其他肝损害因素外所致的以肝实质细胞脂肪变性及贮积为特征的临床病理综合征. 近年来, 随着人们生活方式的改变, NAFLD已成为全球公共健康问题, 其发生率与肥胖、2型糖尿病等代谢综合征相关. 越来越多文献表明肠道菌群与NAFLD的发生发展关系密切: (1)肠道菌群失调可促进宿主吸收更多的能量; (2)肠道菌群失调可诱导机体脂质代谢紊乱, 肝细胞脂质蓄积; (3)肠道菌群失调可增加肠黏膜通透性、促发炎症. 因此, 本文就肠道菌群与NAFLD关系进行整理, 为寻找治疗NAFLD的药物提供新靶点进行简要综述.
Collapse
|
840
|
Turroni S, Brigidi P, Cavalli A, Candela M. Microbiota–Host Transgenomic Metabolism, Bioactive Molecules from the Inside. J Med Chem 2017; 61:47-61. [DOI: 10.1021/acs.jmedchem.7b00244] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Silvia Turroni
- Department
of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro
6, 40126 Bologna, Italy
| | - Patrizia Brigidi
- Department
of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro
6, 40126 Bologna, Italy
| | - Andrea Cavalli
- Department
of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro
6, 40126 Bologna, Italy
- Compunet, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Marco Candela
- Department
of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro
6, 40126 Bologna, Italy
| |
Collapse
|
841
|
Thöni V, Pfister A, Melmer A, Enrich B, Salzmann K, Kaser S, Lamina C, Ebenbichler CF, Hackl H, Tilg H, Moschen AR. Dynamics of Bile Acid Profiles, GLP-1, and FGF19 After Laparoscopic Gastric Banding. J Clin Endocrinol Metab 2017; 102:2974-2984. [PMID: 28591793 DOI: 10.1210/jc.2017-00235] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 06/02/2017] [Indexed: 12/22/2022]
Abstract
CONTEXT An increase of bile acids (BAs), fibroblast growth factor 19 (FGF19), and glucagon-like peptide 1 (GLP-1) has been implicated in metabolic improvements after Roux-en-Y gastric bypass and vertical sleeve gastrectomy. However, data are still conflicting regarding their role after laparoscopic adjustable gastric banding (LAGB). OBJECTIVE To assess the fasting BA, FGF19, and GLP-1 concentrations in plasma before and after LAGB and to test for correlations with immunometabolic parameters. Furthermore, hepatic farnesoid X receptor (FXR) expression and regulation of FXR-dependent genes were analyzed. DESIGN AND SETTING Observational study at the University Hospital Innsbruck. PATIENTS Twenty obese patients. INTERVENTIONS Fasting plasma samples were taken before, 3, 6, and 12 months after LAGB. Liver biopsies were obtained at surgery and after 6 months postoperatively. MAIN OUTCOME MEASURES BA profiles, GLP-1 and FGF19 levels, hepatic FXR expression and regulation of FXR target genes were determined. RESULTS Total, conjugated, and secondary BAs transiently increased 3 months after LAGB (P < 0.01). Only one BA, glycolithocholic acid sulfate, remained significantly elevated throughout the whole follow-up period (P < 0.05). GLP-1 had increased transiently 3 months after surgery (P < 0.01), whereas FGF19 levels increased continuously (P < 0.05). Insulin, homeostasis model assessment index, C-reactive protein, FGF19, and GLP-1 correlated positively with different BAs. No differences were seen in hepatic FXR expression and FXR-regulated genes. CONCLUSIONS Our study results, not only identified LAGB-induced changes in BAs and BA-induced hormones, but also revealed associations between changes in BA profile with GLP-1 and FGF19.
Collapse
Affiliation(s)
- Veronika Thöni
- Department of Medicine, Division of Internal Medicine I (Gastroenterology, Endocrinology, and Metabolism), Medical University of Innsbruck, Innsbruck A-6020, Austria
| | - Alexandra Pfister
- Department of Medicine, Division of Internal Medicine I (Gastroenterology, Endocrinology, and Metabolism), Medical University of Innsbruck, Innsbruck A-6020, Austria
- Christian Doppler Laboratory for Mucosal Immunology, Medical University of Innsbruck, Innsbruck A-6020, Austria
| | - Andreas Melmer
- Department of Medicine, Division of Internal Medicine I (Gastroenterology, Endocrinology, and Metabolism), Medical University of Innsbruck, Innsbruck A-6020, Austria
| | - Barbara Enrich
- Department of Medicine, Division of Internal Medicine I (Gastroenterology, Endocrinology, and Metabolism), Medical University of Innsbruck, Innsbruck A-6020, Austria
| | - Karin Salzmann
- Department of Medicine, Division of Internal Medicine I (Gastroenterology, Endocrinology, and Metabolism), Medical University of Innsbruck, Innsbruck A-6020, Austria
- Christian Doppler Laboratory for Metabolic Crosstalk, Medical University of Innsbruck, Innsbruck A-6020, Austria
| | - Susanne Kaser
- Department of Medicine, Division of Internal Medicine I (Gastroenterology, Endocrinology, and Metabolism), Medical University of Innsbruck, Innsbruck A-6020, Austria
- Christian Doppler Laboratory for Metabolic Crosstalk, Medical University of Innsbruck, Innsbruck A-6020, Austria
| | - Claudia Lamina
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck A-6020, Austria
| | - Christoph F Ebenbichler
- Department of Medicine, Division of Internal Medicine I (Gastroenterology, Endocrinology, and Metabolism), Medical University of Innsbruck, Innsbruck A-6020, Austria
| | - Hubert Hackl
- Biocenter, Division of Bioinformatics, Medical University of Innsbruck, Innsbruck A-6020, Austria
| | - Herbert Tilg
- Department of Medicine, Division of Internal Medicine I (Gastroenterology, Endocrinology, and Metabolism), Medical University of Innsbruck, Innsbruck A-6020, Austria
| | - Alexander R Moschen
- Department of Medicine, Division of Internal Medicine I (Gastroenterology, Endocrinology, and Metabolism), Medical University of Innsbruck, Innsbruck A-6020, Austria
- Christian Doppler Laboratory for Mucosal Immunology, Medical University of Innsbruck, Innsbruck A-6020, Austria
| |
Collapse
|
842
|
Sjöberg BG, Straniero S, Angelin B, Rudling M. Cholestyramine treatment of healthy humans rapidly induces transient hypertriglyceridemia when treatment is initiated. Am J Physiol Endocrinol Metab 2017; 313:E167-E174. [PMID: 28487440 DOI: 10.1152/ajpendo.00416.2016] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 03/29/2017] [Accepted: 05/04/2017] [Indexed: 01/11/2023]
Abstract
Bile acid (BA) production in mice is regulated by hepatic farnesoid X receptors and by intestinal fibroblast growth factor (FGF)-15 (in humans, FGF-19), a suppressor of BA synthesis that also reduces serum triglycerides and glucose. Cholestyramine treatment reduces FGF-19 and induces BA synthesis, whereas plasma triglycerides may increase from unclear reasons. We explored whether FGF-19 may suppress BA synthesis and plasma triglycerides in humans by modulation of FGF-19 levels through long-term cholestyramine treatment at increasing doses. In a second acute experiment, metabolic responses from 1 day of cholestyramine treatment were monitored. Long-term treatment reduced serum FGF-19 by >90%; BA synthesis increased up to 17-fold, whereas serum BAs, triglycerides, glucose, and insulin were stable. After long-term treatment, serum BAs and FGF-19 displayed rebound increases above baseline levels, and BA and cholesterol syntheses normalized after 1 wk without rebound reductions. Acute cholestyramine treatment decreased FGF-19 by 95% overnight and serum BAs by 60%, while BA synthesis increased fourfold and triglycerides doubled. The results support that FGF-19 represses BA synthesis but not serum triglycerides. However, after cessation of both long-term and 1-day cholestyramine treatment, circulating FGF-19 levels were normalized within 2 days, whereas BA synthesis remained significantly induced in both situations, indicating that also other mechanisms than the FGF-19 pathway are responsible for stimulation of BA synthesis elicited by cholestyramine. Several of the responses during cholestyramine treatment persisted at least 6 days after treatment, highlighting the importance of removing such treatment well before evaluating dynamics of the enterohepatic circulation in humans.
Collapse
Affiliation(s)
- Beatrice G Sjöberg
- Metabolism Unit C2:94 and KI/AZ Integrated CardioMetabolic Center, Department of Medicine, and Center for Innovative Medicine, Department of Biosciences and Nutrition, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Sara Straniero
- Metabolism Unit C2:94 and KI/AZ Integrated CardioMetabolic Center, Department of Medicine, and Center for Innovative Medicine, Department of Biosciences and Nutrition, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Bo Angelin
- Metabolism Unit C2:94 and KI/AZ Integrated CardioMetabolic Center, Department of Medicine, and Center for Innovative Medicine, Department of Biosciences and Nutrition, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Mats Rudling
- Metabolism Unit C2:94 and KI/AZ Integrated CardioMetabolic Center, Department of Medicine, and Center for Innovative Medicine, Department of Biosciences and Nutrition, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
| |
Collapse
|
843
|
Albaugh VL, Banan B, Ajouz H, Abumrad NN, Flynn CR. Bile acids and bariatric surgery. Mol Aspects Med 2017; 56:75-89. [PMID: 28390813 PMCID: PMC5603298 DOI: 10.1016/j.mam.2017.04.001] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/27/2017] [Accepted: 04/04/2017] [Indexed: 12/12/2022]
Abstract
Bariatric surgery, specifically Roux-en-Y gastric bypass (RYGB) and vertical sleeve gastrectomy (VSG), are the most effective and durable treatments for morbid obesity and potentially a viable treatment for type 2 diabetes (T2D). The resolution rate of T2D following these procedures is between 40 and 80% and far surpasses that achieved by medical management alone. The molecular basis for this improvement is not entirely understood, but has been attributed in part to the altered enterohepatic circulation of bile acids. In this review we highlight how bile acids potentially contribute to improved lipid and glucose homeostasis, insulin sensitivity and energy expenditure after these procedures. The impact of altered bile acid levels in enterohepatic circulation is also associated with changes in gut microflora, which may further contribute to some of these beneficial effects. We highlight the beneficial effects of experimental surgical procedures in rodents that alter bile secretory flow without gastric restriction or altering nutrient flow. This information suggests a role for bile acids beyond dietary fat emulsification in altering whole body glucose and lipid metabolism strongly, and also suggests emerging roles for the activation of the bile acid receptors farnesoid x receptor (FXR) and G-protein coupled bile acid receptor (TGR5) in these improvements. The limitations of rodent studies and the current state of our understanding is reviewed and the potential effects of bile acids mediating the short- and long-term metabolic improvements after bariatric surgery is critically examined.
Collapse
MESH Headings
- Animals
- Bile Acids and Salts/metabolism
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/microbiology
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/surgery
- Enterohepatic Circulation
- Gastrectomy
- Gastric Bypass
- Gastrointestinal Microbiome/physiology
- Gene Expression Regulation
- Glucose/metabolism
- Homeostasis/physiology
- Humans
- Insulin Resistance
- Obesity, Morbid/metabolism
- Obesity, Morbid/microbiology
- Obesity, Morbid/pathology
- Obesity, Morbid/surgery
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Rodentia
- Signal Transduction
Collapse
Affiliation(s)
- Vance L Albaugh
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Babak Banan
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Hana Ajouz
- American University of Beirut, Beirut, Lebanon
| | - Naji N Abumrad
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Charles R Flynn
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
844
|
Anhê FF, Varin TV, Schertzer JD, Marette A. The Gut Microbiota as a Mediator of Metabolic Benefits after Bariatric Surgery. Can J Diabetes 2017; 41:439-447. [PMID: 28552651 DOI: 10.1016/j.jcjd.2017.02.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 01/25/2017] [Accepted: 02/07/2017] [Indexed: 02/07/2023]
|
845
|
Kim KH, Choi S, Zhou Y, Kim EY, Lee JM, Saha PK, Anakk S, Moore DD. Hepatic FXR/SHP axis modulates systemic glucose and fatty acid homeostasis in aged mice. Hepatology 2017; 66:498-509. [PMID: 28378930 PMCID: PMC8156739 DOI: 10.1002/hep.29199] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 02/25/2017] [Accepted: 03/27/2017] [Indexed: 12/12/2022]
Abstract
UNLABELLED The nuclear receptors farnesoid X receptor (FXR; NR1H4) and small heterodimer partner (SHP; NR0B2) play crucial roles in bile acid homeostasis. Global double knockout of FXR and SHP signaling (DKO) causes severe cholestasis and liver injury at early ages. Here, we report an unexpected beneficial impact on glucose and fatty acid metabolism in aged DKO mice, which show suppressed body weight gain and adiposity when maintained on normal chow. This phenotype was not observed in single Fxr or Shp knockouts. Liver-specific Fxr/Shp double knockout mice fully phenocopied the DKO mice, with lower hepatic triglyceride accumulation, improved glucose/insulin tolerance, and accelerated fatty acid use. In both DKO and liver-specific Fxr/Shp double knockout livers, these metabolic phenotypes were associated with altered expression of fatty acid metabolism and autophagy-machinery genes. Loss of the hepatic FXR/SHP axis reprogrammed white and brown adipose tissue gene expression to boost fatty acid usage. CONCLUSION Combined deletion of the hepatic FXR/SHP axis improves glucose/fatty acid homeostasis in aged mice, reversing the aging phenotype of body weight gain, increased adiposity, and glucose/insulin tolerance, suggesting a central role of this axis in whole-body energy homeostasis. (Hepatology 2017;66:498-509).
Collapse
Affiliation(s)
- Kang Ho Kim
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - Sungwoo Choi
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX
| | - Ying Zhou
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX,Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, TX
| | - Eun Young Kim
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jae Man Lee
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Pradip K. Saha
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - Sayeepriyadarshini Anakk
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL
| | - David D. Moore
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX,Program in Developmental Biology, Baylor College of Medicine, Houston, TX,Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, TX
| |
Collapse
|
846
|
Li R, Wang H, Shi Q, Wang N, Zhang Z, Xiong C, Liu J, Chen Y, Jiang L, Jiang Q. Effects of oral florfenicol and azithromycin on gut microbiota and adipogenesis in mice. PLoS One 2017; 12:e0181690. [PMID: 28742883 PMCID: PMC5526540 DOI: 10.1371/journal.pone.0181690] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 07/04/2017] [Indexed: 12/11/2022] Open
Abstract
Certain antibiotics detected in urine are associated with childhood obesity. In the current experimental study, we investigated two representative antibiotics detected in urine, florfenicol and azithromycin, for their early effects on adipogenesis, gut microbiota, short-chain fatty acids (SCFAs), and bile acids in mice. Thirty C57BL/6 mice aged four weeks were randomly divided into three groups (florfenicol, azithromycin and control). The two experimental groups were administered florfenicol or azithromycin at 5 mg/kg/day for four weeks. Body weight was measured weekly. The composition of the gut microbiota, body fat, SCFAs, and bile acids in colon contents were measured at the end of the experiment. The composition of the gut microbiota was determined by sequencing the bacterial 16S rRNA gene. The concentration of SCFAs and bile acids was determined using gas chromatography and liquid chromatography coupled to tandem mass spectrometry, respectively. The composition of the gut microbiota indicated that the two antibiotics altered the gut microbiota composition and decreased its richness and diversity. At the phylum level, the ratio of Firmicutes/Bacteroidetes increased significantly in the antibiotic groups. At the genus level, there were declines in Christensenella, Gordonibacter and Anaerotruncus in the florfenicol group, in Lactobacillus in the azithromycin group, and in Alistipes, Desulfovibrio, Parasutterella and Rikenella in both the antibiotic groups. The decrease in Rikenella in the azithromycin group was particularly noticeable. The concentration of SCFAs and secondary bile acids decreased in the colon, but the concentration of primary bile acids increased. These findings indicated that florfenicol and azithromycin increased adipogenesis and altered gut microbiota composition, SCFA production, and bile acid metabolism, suggesting that exposure to antibiotics might be one risk factor for childhood obesity. More studies are needed to investigate the specific mechanisms.
Collapse
Affiliation(s)
- Rui Li
- Key Laboratory of Public Health Safety of Ministry of Education, School of Public Health, Fudan University, Shanghai, China
| | - Hexing Wang
- Key Laboratory of Public Health Safety of Ministry of Education, School of Public Health, Fudan University, Shanghai, China
| | - Qingfeng Shi
- Key Laboratory of Public Health Safety of Ministry of Education, School of Public Health, Fudan University, Shanghai, China
| | - Na Wang
- Key Laboratory of Public Health Safety of Ministry of Education, School of Public Health, Fudan University, Shanghai, China
| | - Zhijie Zhang
- Key Laboratory of Public Health Safety of Ministry of Education, School of Public Health, Fudan University, Shanghai, China
| | - Chenglong Xiong
- Key Laboratory of Public Health Safety of Ministry of Education, School of Public Health, Fudan University, Shanghai, China
| | - Jianxiang Liu
- Key Laboratory of Public Health Safety of Ministry of Education, School of Public Health, Fudan University, Shanghai, China
| | - Yue Chen
- School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Lufang Jiang
- Key Laboratory of Public Health Safety of Ministry of Education, School of Public Health, Fudan University, Shanghai, China
- * E-mail:
| | - Qingwu Jiang
- Key Laboratory of Public Health Safety of Ministry of Education, School of Public Health, Fudan University, Shanghai, China
| |
Collapse
|
847
|
Diet-induced obesity and weight loss alter bile acid concentrations and bile acid-sensitive gene expression in insulin target tissues of C57BL/6J mice. Nutr Res 2017; 46:11-21. [PMID: 29173647 DOI: 10.1016/j.nutres.2017.07.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 06/13/2017] [Accepted: 07/20/2017] [Indexed: 12/31/2022]
Abstract
Bile acids (BAs) influence the metabolism of glucose, lipids, and energy expenditure. We hypothesized that BA concentrations and related gene expression would be altered in lean (low-fat diet fed; LFD) vs diet-induced obese (high-fat diet fed; HFD) groups of mice and that some detected changes would remain after weight loss in an HFD group switched to the LFD (SW). Taurine conjugates dominated the bile acid composition of the liver, epididymal white adipose tissue (eWAT), and hypothalamus, with the latter having lower levels (~95%, ~95%, and ~80%, respectively; P<.05). Plasma conjugated bile acids were elevated in the HFD relative to the LFD and SW animals. Total hepatic BA concentrations decreased in obese mice fed HFD, and levels returned to preobese levels in the SW group. Subtle changes in unconjugated bile acids were detected in the eWAT, hypothalamus, and muscle. Liver expression of a variety of enzymes involved in BA synthesis (eg, Cyp27a1, Acox2), BA transport (eg, Slc22a8), and BA-sensitive receptors (Fxr, Tgr5) were unchanged by HFD feeding but decreased with SW. Other hepatic enzymes were induced in the SW group (eg, Amacr and Bal). In eWAT, Cyp27a1 and Acox2 also declined in the SW group, whereas the HFD group showed reduced expression of BA transporters (eg, Abcc3), and changes in Fxr and Tgr5 were unclear. Therefore, although most detectable changes in BA metabolism associated with diet-induced obesity are reversed by diet-induced weight loss, some effects on BA composition, concentrations, and gene expression can persist after weight loss.
Collapse
|
848
|
Lowered fasting chenodeoxycholic acid correlated with the decrease of fibroblast growth factor 19 in Chinese subjects with impaired fasting glucose. Sci Rep 2017; 7:6042. [PMID: 28729691 PMCID: PMC5519593 DOI: 10.1038/s41598-017-06252-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 06/12/2017] [Indexed: 12/17/2022] Open
Abstract
The gut-derived hormone Fibroblast growth factor 19 (FGF19) could regulate glucose metabolism and is induced by bile acids (BAs) through activating Farnesoid X Receptor (FXR). FGF19 was found to decrease in subjects with isolated-impaired fasting glucose (I-IFG) and type 2 diabetes mellitus (T2DM). However, the reason for the change of FGF19 in subjects with different glucometabolic status remained unclear. Here we measured six BAs including chenodeoxycholic acid (CDCA), cholic acid, deoxycholic acid, their glycine conjugates and FGF19 levels during oral glucose tolerance test (OGTT) in normal glucose tolerance (NGT), isolated-impaired glucose tolerance, I-IFG, combined glucose intolerance (CGI) and T2DM subjects. After OGTT, serum FGF19 peaked at 120 min in all subjects. Glycine conjugated BAs peaked at 30 min, while free BAs did not elevated significantly. Consistent with the decrease trend in FGF19 levels, fasting serum CDCA levels in subjects with I-IFG, CGI and T2DM were significantly lower than NGT subjects (P < 0.05). Fasting serum CDCA was independently associated with FGF19. CDCA strongly upregulated FGF19 mRNA levels in LS174T cells in a dose- and time-dependent manner. These results suggest that the decrease of FGF19 in subjects with I-IFG was at least partially due to their decrease of CDCA acting via FXR.
Collapse
|
849
|
肖 叔, 于 珮. 肠道菌群与2型糖尿病及并发症. Shijie Huaren Xiaohua Zazhi 2017; 25:1819-1831. [DOI: 10.11569/wcjd.v25.i20.1819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
2型糖尿病(type 2 diabetes mellitus, T2DM)是一种由多因素导致的代谢性疾病, 关于其发病机制的研究逐渐深入, 其中肠道菌群与糖尿病的关系成为近年来研究的焦点. 肠道菌群与人体健康息息相关, 越来越多的研究发现肠道菌群的失调可能通过多种机制参与T2DM的发生与发展, 而T2DM病患者体内代谢环境的改变又可能会影响肠道菌群的组成和功能. 在这篇综述中, 我们总结最近的相关研究探讨肠道菌群失调在T2DM发生和发展中的作用、有关的治疗策略以及未来的研究前景.
Collapse
|
850
|
Ladurner A, Zehl M, Grienke U, Hofstadler C, Faur N, Pereira FC, Berry D, Dirsch VM, Rollinger JM. Allspice and Clove As Source of Triterpene Acids Activating the G Protein-Coupled Bile Acid Receptor TGR5. Front Pharmacol 2017; 8:468. [PMID: 28769799 PMCID: PMC5511840 DOI: 10.3389/fphar.2017.00468] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 06/30/2017] [Indexed: 12/31/2022] Open
Abstract
Worldwide, metabolic diseases such as obesity and type 2 diabetes have reached epidemic proportions. A major regulator of metabolic processes that gained interest in recent years is the bile acid receptor TGR5 (Takeda G protein-coupled receptor 5). This G protein-coupled membrane receptor can be found predominantly in the intestine, where it is mainly responsible for the secretion of the incretins glucagon-like peptide 1 (GLP-1) and peptide YY (PYY). The aim of this study was (i) to identify plant extracts with TGR5-activating potential, (ii) to narrow down their activity to the responsible constituents, and (iii) to assess whether the intestinal microbiota produces transformed metabolites with a different activity profile. Chenodeoxycholic acid (CDCA) served as positive control for both, the applied cell-based luciferase reporter gene assay for TGR5 activity and the biotransformation assay using mouse fecal slurry. The suitability of the workflow was demonstrated by the biotransformation of CDCA to lithocholic acid resulting in a distinct increase in TGR5 activity. Based on a traditional Tibetan formula, 19 plant extracts were selected and investigated for TGR5 activation. Extracts from the commonly used spices Syzygium aromaticum (SaroE, clove), Pimenta dioica (PdioE, allspice), and Kaempferia galanga (KgalE, aromatic ginger) significantly increased TGR5 activity. After biotransformation, only KgalE showed significant differences in its metabolite profile, which, however, did not alter its TGR5 activity compared to non-transformed KgalE. UHPLC-HRMS (high-resolution mass spectrometry) analysis revealed triterpene acids (TTAs) as the main constituents of the extracts SaroE and PdioE. Identification and quantification of TTAs in these two extracts as well as comparison of their TGR5 activity with reconstituted TTA mixtures allowed the attribution of the TGR5 activity to TTAs. EC50s were determined for the main TTAs, i.e., oleanolic acid (2.2 ± 1.6 μM), ursolic acid (1.1 ± 0.2 μM), as well as for the hitherto unknown TGR5 activators corosolic acid (0.5 ± 1.0 μM) and maslinic acid (3.7 ± 0.7 μM). In conclusion, extracts of clove, allspice, and aromatic ginger activate TGR5, which might play a pivotal role in their therapeutic use for the treatment of metabolic diseases. Moreover, the TGR5 activation of SaroE and PdioE could be pinpointed solely to TTAs.
Collapse
Affiliation(s)
- Angela Ladurner
- Department of Pharmacognosy, Faculty of Life Sciences, University of ViennaVienna, Austria
| | - Martin Zehl
- Department of Pharmacognosy, Faculty of Life Sciences, University of ViennaVienna, Austria
- Department of Pharmaceutical Chemistry, University of ViennaVienna, Austria
- Department of Analytical Chemistry, University of ViennaVienna, Austria
| | - Ulrike Grienke
- Department of Pharmacognosy, Faculty of Life Sciences, University of ViennaVienna, Austria
| | - Christoph Hofstadler
- Department of Pharmacognosy, Faculty of Life Sciences, University of ViennaVienna, Austria
| | - Nadina Faur
- Department of Pharmacognosy, Faculty of Life Sciences, University of ViennaVienna, Austria
| | - Fátima C. Pereira
- Department of Microbiology and Ecosystem Science, University of ViennaVienna, Austria
| | - David Berry
- Department of Microbiology and Ecosystem Science, University of ViennaVienna, Austria
| | - Verena M. Dirsch
- Department of Pharmacognosy, Faculty of Life Sciences, University of ViennaVienna, Austria
| | - Judith M. Rollinger
- Department of Pharmacognosy, Faculty of Life Sciences, University of ViennaVienna, Austria
| |
Collapse
|