1001
|
Yuan L, Bambha K. Bile acid receptors and nonalcoholic fatty liver disease. World J Hepatol 2015; 7:2811-2818. [PMID: 26668692 PMCID: PMC4670952 DOI: 10.4254/wjh.v7.i28.2811] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 09/14/2015] [Accepted: 11/25/2015] [Indexed: 02/06/2023] Open
Abstract
With the high prevalence of obesity, diabetes, and other features of the metabolic syndrome in United States, nonalcoholic fatty liver disease (NAFLD) has inevitably become a very prevalent chronic liver disease and is now emerging as one of the leading indications for liver transplantation. Insulin resistance and derangement of lipid metabolism, accompanied by activation of the pro-inflammatory response and fibrogenesis, are essential pathways in the development of the more clinically significant form of NAFLD, known as nonalcoholic steatohepatitis (NASH). Recent advances in the functional characterization of bile acid receptors, such as farnesoid X receptor (FXR) and transmembrane G protein-coupled receptor (TGR) 5, have provided further insight in the pathophysiology of NASH and have led to the development of potential therapeutic targets for NAFLD and NASH. Beyond maintaining bile acid metabolism, FXR and TGR5 also regulate lipid metabolism, maintain glucose homeostasis, increase energy expenditure, and ameliorate hepatic inflammation. These intriguing features have been exploited to develop bile acid analogues to target pathways in NAFLD and NASH pathogenesis. This review provides a brief overview of the pathogenesis of NAFLD and NASH, and then delves into the biological functions of bile acid receptors, particularly with respect to NASH pathogenesis, with a description of the associated experimental data, and, finally, we discuss the prospects of bile acid analogues in the treatment of NAFLD and NASH.
Collapse
|
1002
|
Miele L, Giorgio V, Alberelli MA, De Candia E, Gasbarrini A, Grieco A. Impact of Gut Microbiota on Obesity, Diabetes, and Cardiovascular Disease Risk. Curr Cardiol Rep 2015; 17:120. [PMID: 26497040 DOI: 10.1007/s11886-015-0671-z] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Gut microbiota has been recently established to have a contributory role in the development of cardiometabolic disorders, such as atherosclerosis, obesity, and type 2 diabetes. Growing interest has focused on the modulation of gut microbiota as a therapeutic strategy in cardiovascular diseases and metabolic disorders. In this paper, we have reviewed the impact of gut microbiota on metabolic disorders and cardiovascular disease risk, focusing on the newest findings in this field.
Collapse
Affiliation(s)
- Luca Miele
- Institute of Internal Medicine, Policlinico Gemelli Hospital, Catholic University of Sacred Heart of Rome, Rome, Italy.
- Clinical Division of Internal Medicine, Gastroenterology and Liver Unit, Complesso Integrato Columbus Hospital, Catholic University of Sacred Heart of Rome, Rome, Italy.
| | - Valentina Giorgio
- Pediatric Department, Policlinico Gemelli Hospital, Catholic University of Sacred Heart of Rome, Rome, Italy.
| | - Maria Adele Alberelli
- Institute of Internal Medicine, Policlinico Gemelli Hospital, Catholic University of Sacred Heart of Rome, Rome, Italy.
| | - Erica De Candia
- Institute of Internal Medicine, Policlinico Gemelli Hospital, Catholic University of Sacred Heart of Rome, Rome, Italy.
| | - Antonio Gasbarrini
- Institute of Internal Medicine, Policlinico Gemelli Hospital, Catholic University of Sacred Heart of Rome, Rome, Italy.
| | - Antonio Grieco
- Institute of Internal Medicine, Policlinico Gemelli Hospital, Catholic University of Sacred Heart of Rome, Rome, Italy.
| |
Collapse
|
1003
|
Chai J, Zou L, Li X, Han D, Wang S, Hu S, Guan J. Mechanism of bile acid-regulated glucose and lipid metabolism in duodenal-jejunal bypass. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:15778-15785. [PMID: 26884847 PMCID: PMC4730060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 10/25/2015] [Indexed: 06/05/2023]
Abstract
Bile acid plays an important role in regulating blood glucose, lipid and energy metabolism. The present study was implemented to determine the effect of duodenal-jejunal bypass (DJB) on FXR, TGR-5expression in terminal ileum and its bile acid-related mechanism on glucose and lipid metabolism. Immunohistochemistry was used to detect relative gene or protein expression in liver and intestine. Firstly, we found that expression of FXR in liver and terminal ileum of DJB group was significantly higher than that in S-DJB group (P<0.05). In addition, DJB dramatically increased the activation of TGR-5 in the liver of rats. Furthermore, PEPCK, G6Pase, FBPase 1 and GLP-1 were up-regulated by DJB. In conclusion, these results showed that bile acid ameliorated glucose and lipid metabolism through bile acid-FXR and bile acid- TGR-5 signaling pathway.
Collapse
Affiliation(s)
- Jie Chai
- Department of General Surgery, Shandong Cancer Hospital and InstituteJinan 250117, China
| | - Lei Zou
- Department of General Surgery, Shandong Cancer Hospital and InstituteJinan 250117, China
| | - Xirui Li
- Department of General Surgery, Shandong Cancer Hospital and InstituteJinan 250117, China
| | - Dali Han
- Department of General Surgery, Shandong Cancer Hospital and InstituteJinan 250117, China
| | - Shan Wang
- Department of General Surgery, Shandong Cancer Hospital and InstituteJinan 250117, China
| | - Sanyuan Hu
- Department of General Surgery, Qilu Hospital of Shandong UniversityJinan 250012, China
| | - Jie Guan
- Department of General Surgery, Shandong Cancer Hospital and InstituteJinan 250117, China
| |
Collapse
|
1004
|
Cole AJ, Teigen LM, Jahansouz C, Earthman CP, Sibley SD. The Influence of Bariatric Surgery on Serum Bile Acids in Humans and Potential Metabolic and Hormonal Implications: a Systematic Review. Curr Obes Rep 2015; 4:441-50. [PMID: 26335653 DOI: 10.1007/s13679-015-0171-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recent research suggests a mechanistic role for bile acids (BA) in the metabolic improvement following bariatric surgery. It is believed that the hormonal and metabolic effects associated with changes in systemic BAs may be related to the farnesoid X receptor (FXR) and a G-protein coupled receptor (TGR5). This systematic review examines changes in systemic BAs following bariatric procedures. Studies were included if they reported the measurement of systemic BAs in humans at at least one time point after bariatric surgery. Eleven papers were identified that met the inclusion criteria. Seven studies reported the effect of Roux-en-Y gastric bypass (RYGB) on fasting BAs. The majority (6/7) reported that fasting BAs increased after RYGB. Data regarding fasting BAs after vertical sleeve gastrectomy (VSG) and laparoscopic gastric banding (LAGB) are inconsistent. Data regarding post-prandial BA changes after RYGB, VSG, and LAGB are also inconsistent. More research is needed to investigate the connection between BAs and the metabolic improvement seen after bariatric surgery.
Collapse
Affiliation(s)
- Abigail J Cole
- Department of Food Science and Nutrition, University of Minnesota-Twin Cities, 1334 Eckles Avenue, Saint Paul, MN, 55108, USA.
| | - Levi M Teigen
- Department of Food Science and Nutrition, University of Minnesota-Twin Cities, 1334 Eckles Avenue, Saint Paul, MN, 55108, USA.
| | - Cyrus Jahansouz
- Medical School, Department of Surgery, University of Minnesota-Twin Cities, 420 Delaware Street SE, Minneapolis, MN, 55404, USA.
| | - Carrie P Earthman
- Department of Food Science and Nutrition, University of Minnesota-Twin Cities, 1334 Eckles Avenue, Saint Paul, MN, 55108, USA.
| | - Shalamar D Sibley
- Department of Medicine, Division of Endocrinology, University of Minnesota-Twin Cities, 516 Delaware St. SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
1005
|
Lee EY, Sakurai K, Zhang X, Toda C, Tanaka T, Jiang M, Shirasawa T, Tachibana K, Yokote K, Vidal-Puig A, Minokoshi Y, Miki T. Unsuppressed lipolysis in adipocytes is linked with enhanced gluconeogenesis and altered bile acid physiology in Insr(P1195L/+) mice fed high-fat-diet. Sci Rep 2015; 5:17565. [PMID: 26615883 PMCID: PMC4663474 DOI: 10.1038/srep17565] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 11/02/2015] [Indexed: 12/31/2022] Open
Abstract
High-fat diet (HFD) triggers insulin resistance and diabetes mellitus, but their link remains unclear. Characterization of overt hyperglycemia in insulin receptor mutant (Insr(P1195L/+)) mice exposed to HFD (Insr(P1195L/+)/HFD mice) revealed increased glucose-6-phosphatase (G6pc) expression in liver and increased gluconeogenesis from glycerol. Lipolysis in white adipose tissues (WAT) and lipolysis-induced blood glucose rise were increased in Insr(P1195L/+)/HFD mice, while wild-type WAT transplantation ameliorated the hyperglycemia and the increased G6pc expression. We found that the expressions of genes involved in bile acid (BA) metabolism were altered in Insr(P1195L/+)/HFD liver. Among these, the expression of Cyp7a1, a BA synthesis enzyme, was insulin-dependent and was markedly decreased in Insr(P1195L/+)/HFD liver. Reduced Cyp7a1 expression in Insr(P1195L/+)/HFD liver was rescued by WAT transplantation, and the expression of Cyp7a1 was suppressed by glycerol administration in wild-type liver. These findings suggest that unsuppressed lipolysis in adipocytes elicited by HFD feeding is linked with enhanced gluconeogenesis from glycerol and with alterations in BA physiology in Insr(P1195L/+)/HFD liver.
Collapse
Affiliation(s)
- Eun Young Lee
- Department of Medical Physiology, Chiba University, Graduate School of Medicine, Chiba 260-8670 Japan
| | - Kenichi Sakurai
- Department of Clinical Cell Biology and Medicine, Chiba University, Graduate School of Medicine, Chiba 260-8670 Japan
| | - Xilin Zhang
- Department of Medical Physiology, Chiba University, Graduate School of Medicine, Chiba 260-8670 Japan
| | - Chitoku Toda
- Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| | - Tomoaki Tanaka
- Department of Clinical Cell Biology and Medicine, Chiba University, Graduate School of Medicine, Chiba 260-8670 Japan
| | - Meizi Jiang
- Department of Genome Research and Clinical Application, Chiba University, Graduate School of Medicine, Chiba 260-8670 Japan
| | - Takuji Shirasawa
- Department of Ageing Control, Juntendo University, Graduate School of Medicine. Bunkyo 113-0033, Japan
| | - Kaori Tachibana
- Department of Clinical Cell Biology and Medicine, Chiba University, Graduate School of Medicine, Chiba 260-8670 Japan
| | - Koutaro Yokote
- Department of Clinical Cell Biology and Medicine, Chiba University, Graduate School of Medicine, Chiba 260-8670 Japan
| | - Antonio Vidal-Puig
- Department of Clinical Biochemistry, Metabolic Research Laboratories, Addenbrooke's Treatment Centre, Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Yasuhiko Minokoshi
- Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| | - Takashi Miki
- Department of Medical Physiology, Chiba University, Graduate School of Medicine, Chiba 260-8670 Japan
| |
Collapse
|
1006
|
Zietek T, Rath E, Haller D, Daniel H. Intestinal organoids for assessing nutrient transport, sensing and incretin secretion. Sci Rep 2015; 5:16831. [PMID: 26582215 PMCID: PMC4652176 DOI: 10.1038/srep16831] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 10/21/2015] [Indexed: 12/31/2022] Open
Abstract
Intestinal nutrient transport and sensing are of emerging interest in research on obesity and diabetes and as drug targets. Appropriate in vitro models are lacking that allow both, studies on transport processes as well as sensing and subsequent incretin hormone secretion including intracellular signaling. We here demonstrate that murine small-intestinal organoids are the first in vitro model system enabling concurrent investigations of nutrient and drug transport, sensing and incretin hormone secretion as well as fluorescent live-cell imaging of intracellular signaling processes. By generating organoid cultures from wild type mice and animals lacking different nutrient transporters, we show that organoids preserve the main phenotypic features and functional characteristics of the intestine. This turns them into the best in vitro model currently available and opens new avenues for basic as well as medical research.
Collapse
Affiliation(s)
- Tamara Zietek
- Department of Nutritional Physiology, Technische Universität München, 85350 Freising, Germany.,ZIEL-Institute for Food &Health, 85350 Freising, Germany
| | - Eva Rath
- ZIEL-Institute for Food &Health, 85350 Freising, Germany.,Chair of Nutrition and Immunology, Technische Universität München, 85350 Freising, Germany
| | - Dirk Haller
- ZIEL-Institute for Food &Health, 85350 Freising, Germany.,Chair of Nutrition and Immunology, Technische Universität München, 85350 Freising, Germany
| | - Hannelore Daniel
- Department of Nutritional Physiology, Technische Universität München, 85350 Freising, Germany.,ZIEL-Institute for Food &Health, 85350 Freising, Germany
| |
Collapse
|
1007
|
Cavalcante-Silva LHA, Galvão JGFM, da Silva JSDF, de Sales-Neto JM, Rodrigues-Mascarenhas S. Obesity-Driven Gut Microbiota Inflammatory Pathways to Metabolic Syndrome. Front Physiol 2015; 6:341. [PMID: 26635627 PMCID: PMC4652019 DOI: 10.3389/fphys.2015.00341] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 11/03/2015] [Indexed: 12/28/2022] Open
Abstract
The intimate interplay between immune system, metabolism, and gut microbiota plays an important role in controlling metabolic homeostasis and possible obesity development. Obesity involves impairment of immune response affecting both innate and adaptive immunity. The main factors involved in the relationship of obesity with inflammation have not been completely elucidated. On the other hand, gut microbiota, via innate immune receptors, has emerged as one of the key factors regulating events triggering acute inflammation associated with obesity and metabolic syndrome. Inflammatory disorders lead to several signaling transduction pathways activation, inflammatory cytokine, chemokine production and cell migration, which in turn cause metabolic dysfunction. Inflamed adipose tissue, with increased macrophages infiltration, is associated with impaired preadipocyte development and differentiation to mature adipose cells, leading to ectopic lipid accumulation and insulin resistance. This review focuses on the relationship between obesity and inflammation, which is essential to understand the pathological mechanisms governing metabolic syndrome.
Collapse
Affiliation(s)
- Luiz H A Cavalcante-Silva
- Programa Multicêntrico de Pós-graduação em Ciências Fisiológicas, Laboratório de Imunofarmacologia, Centro de Biotecnologia, Universidade Federal da Paraíba João Pessoa, Brasil
| | - José G F M Galvão
- Programa Multicêntrico de Pós-graduação em Ciências Fisiológicas, Laboratório de Imunofarmacologia, Centro de Biotecnologia, Universidade Federal da Paraíba João Pessoa, Brasil
| | - Juliane Santos de França da Silva
- Programa de Pós-Graduação em Produtos Naturais e Sintéticos Bioativos, Laboratório de Imunofarmacologia, Centro de Ciências da Saúde, Universidade Federal da Paraíba João Pessoa, Brasil
| | - José M de Sales-Neto
- Programa de Pós-Graduação em Biotecnologia, Laboratório de Imunofarmacologia, Centro de Biotecnologia, Universidade Federal da Paraíba João Pessoa, Brasil
| | - Sandra Rodrigues-Mascarenhas
- Programa Multicêntrico de Pós-graduação em Ciências Fisiológicas, Laboratório de Imunofarmacologia, Centro de Biotecnologia, Universidade Federal da Paraíba João Pessoa, Brasil ; Programa de Pós-Graduação em Produtos Naturais e Sintéticos Bioativos, Laboratório de Imunofarmacologia, Centro de Ciências da Saúde, Universidade Federal da Paraíba João Pessoa, Brasil ; Programa de Pós-Graduação em Biotecnologia, Laboratório de Imunofarmacologia, Centro de Biotecnologia, Universidade Federal da Paraíba João Pessoa, Brasil
| |
Collapse
|
1008
|
Di Leva FS, Festa C, Renga B, Sepe V, Novellino E, Fiorucci S, Zampella A, Limongelli V. Structure-based drug design targeting the cell membrane receptor GPBAR1: exploiting the bile acid scaffold towards selective agonism. Sci Rep 2015; 5:16605. [PMID: 26567894 PMCID: PMC4645117 DOI: 10.1038/srep16605] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 10/16/2015] [Indexed: 12/21/2022] Open
Abstract
Bile acids can regulate nutrient metabolism through the activation of the cell membrane receptor GPBAR1 and the nuclear receptor FXR. Developing an exogenous control over these receptors represents an attractive strategy for the treatment of enterohepatic and metabolic disorders. A number of dual GPBAR1/FXR agonists are known, however their therapeutic use is limited by multiple unwanted effects due to activation of the diverse downstream signals controlled by the two receptors. On the other hand, designing selective GPBAR1 and FXR agonists is challenging since the two proteins share similar structural requisites for ligand binding. Here, taking advantage of our knowledge of the two targets, we have identified through a rational drug design study a series of amine lithocholic acid derivatives as selective GPBAR1 agonists. The presence of the 3α-NH2 group on the steroidal scaffold is responsible for the selectivity over FXR unveiling unprecedented structural insights into bile acid receptors activity modulation.
Collapse
Affiliation(s)
- Francesco Saverio Di Leva
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, I-80131 Naples, Italy
| | - Carmen Festa
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, I-80131 Naples, Italy
| | - Barbara Renga
- Department of Surgery and Biomedical Sciences, Nuova Facoltà di Medicina, P.zza L. Severi, I-06132 Perugia, Italy
| | - Valentina Sepe
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, I-80131 Naples, Italy
| | - Ettore Novellino
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, I-80131 Naples, Italy
| | - Stefano Fiorucci
- Department of Surgery and Biomedical Sciences, Nuova Facoltà di Medicina, P.zza L. Severi, I-06132 Perugia, Italy
| | - Angela Zampella
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, I-80131 Naples, Italy
| | - Vittorio Limongelli
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, I-80131 Naples, Italy.,Università della Svizzera Italiana (USI), Faculty of Informatics, Institute of Computational Science, via G. Buffi 13, CH-6900 Lugano, Switzerland
| |
Collapse
|
1009
|
Carotti S, Guarino MPL, Vespasiani-Gentilucci U, Morini S. Starring role of toll-like receptor-4 activation in the gut-liver axis. World J Gastrointest Pathophysiol 2015; 6:99-109. [PMID: 26600967 PMCID: PMC4644892 DOI: 10.4291/wjgp.v6.i4.99] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 07/21/2015] [Accepted: 10/19/2015] [Indexed: 02/06/2023] Open
Abstract
Since the introduction of the term “gut-liver axis”, many studies have focused on the functional links of intestinal microbiota, barrier function and immune responses to liver physiology. Intestinal and extra-intestinal diseases alter microbiota composition and lead to dysbiosis, which aggravates impaired intestinal barrier function via increased lipopolysaccharide translocation. The subsequent increased passage of gut-derived product from the intestinal lumen to the organ wall and bloodstream affects gut motility and liver biology. The activation of the toll-like receptor 4 (TLR-4) likely plays a key role in both cases. This review analyzed the most recent literature on the gut-liver axis, with a particular focus on the role of TLR-4 activation. Findings that linked liver disease with dysbiosis are evaluated, and links between dysbiosis and alterations of intestinal permeability and motility are discussed. We also examine the mechanisms of translocated gut bacteria and/or the bacterial product activation of liver inflammation and fibrogenesis via activity on different hepatic cell types.
Collapse
|
1010
|
TGR5 and Immunometabolism: Insights from Physiology and Pharmacology. Trends Pharmacol Sci 2015; 36:847-857. [PMID: 26541439 DOI: 10.1016/j.tips.2015.08.002] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 08/02/2015] [Accepted: 08/04/2015] [Indexed: 12/15/2022]
Abstract
In the past decade substantial progress has been made in understanding how the insurgence of chronic low-grade inflammation influences the physiology of several metabolic diseases. Tissue-resident immune cells have been identified as central players in these processes, linking inflammation to metabolism. The bile acid-responsive G-protein-coupled receptor TGR5 is expressed in monocytes and macrophages, and its activation mediates potent anti-inflammatory effects. Herein, we summarize recent advances in TGR5 research, focusing on the downstream effector pathways that are modulated by TGR5 activators, and on its therapeutic potential in inflammatory and metabolic diseases.
Collapse
|
1011
|
Qian LL, Li HT, Zhang L, Fang QC, Jia WP. Effect of the Gut Microbiota on Obesity and Its Underlying Mechanisms: an Update. BIOMEDICAL AND ENVIRONMENTAL SCIENCES : BES 2015; 28:839-847. [PMID: 26695364 DOI: 10.1016/s0895-3988(15)30116-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Accepted: 10/27/2015] [Indexed: 01/05/2025]
Affiliation(s)
- Ling Ling Qian
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai 200025, China; Department of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hua Ting Li
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai 200025, China
| | - Lei Zhang
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai 200025, China; Department of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qi Chen Fang
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai 200025, China
| | - Wei Ping Jia
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai 200025, China; Department of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
1012
|
Ferslew BC, Xie G, Johnston CK, Su M, Stewart PW, Jia W, Brouwer KLR, Barritt AS. Altered Bile Acid Metabolome in Patients with Nonalcoholic Steatohepatitis. Dig Dis Sci 2015; 60:3318-3328. [PMID: 26138654 PMCID: PMC4864493 DOI: 10.1007/s10620-015-3776-8] [Citation(s) in RCA: 268] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 06/17/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS The prevalence of nonalcoholic fatty liver disease (NAFLD) and steatohepatitis (NASH) is increasing at an alarming rate. The role of bile acids in the development and progression of NAFLD to NASH and cirrhosis is poorly understood. This study aimed to quantify the bile acid metabolome in healthy subjects and patients with non-cirrhotic NASH under fasting conditions and after a standardized meal. METHODS Liquid chromatography tandem mass spectroscopy was used to quantify 30 serum and 16 urinary bile acids from 15 healthy volunteers and 7 patients with biopsy-confirmed NASH. Bile acid concentrations were measured at two fasting and four post-prandial time points following a high-fat meal to induce gallbladder contraction and bile acid reabsorption from the intestine. RESULTS Patients with NASH had significantly higher total serum bile acid concentrations than healthy subjects under fasting conditions (2.2- to 2.4-fold increase in NASH; NASH 2595-3549 µM and healthy 1171-1458 µM) and at all post-prandial time points (1.7- to 2.2-fold increase in NASH; NASH 4444-5898 µM and healthy 2634-2829 µM). These changes were driven by increased taurine- and glycine-conjugated primary and secondary bile acids. Patients with NASH exhibited greater variability in their fasting and post-prandial bile acid profile. CONCLUSIONS Results indicate that patients with NASH have higher fasting and post-prandial exposure to bile acids, including the more hydrophobic and cytotoxic secondary species. Increased bile acid exposure may be involved in liver injury and the pathogenesis of NAFLD and NASH.
Collapse
Affiliation(s)
- Brian C Ferslew
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Kerr Hall, CB #7569, Chapel Hill, NC, 27599, USA.
- Clinical Pharmacology and Drug Metabolism and Pharmacokinetics, Theravance Biopharma US, Inc., 901 Gateway Blvd, South San Francisco, CA, 94080, USA.
| | - Guoxiang Xie
- Metabolomics Shared Resource, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA.
| | - Curtis K Johnston
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Kerr Hall, CB #7569, Chapel Hill, NC, 27599, USA.
| | - Mingming Su
- Metabolomics Shared Resource, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA.
| | - Paul W Stewart
- Department of Biostatistics, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, 3105G McGavran-Greenberg Hall, Chapel Hill, NC, 27599, USA.
| | - Wei Jia
- Metabolomics Shared Resource, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA.
| | - Kim L R Brouwer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Kerr Hall, CB #7569, Chapel Hill, NC, 27599, USA.
| | - A Sidney Barritt
- Division of Gastroenterology and Hepatology, UNC School of Medicine, University of North Carolina at Chapel Hill, 8004 Burnett Womack, CB #7584, Chapel Hill, NC, 27599-7584, USA.
| |
Collapse
|
1013
|
Brighton CA, Rievaj J, Kuhre RE, Glass LL, Schoonjans K, Holst JJ, Gribble FM, Reimann F. Bile Acids Trigger GLP-1 Release Predominantly by Accessing Basolaterally Located G Protein-Coupled Bile Acid Receptors. Endocrinology 2015; 156:3961-70. [PMID: 26280129 PMCID: PMC4606749 DOI: 10.1210/en.2015-1321] [Citation(s) in RCA: 255] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Bile acids are well-recognized stimuli of glucagon-like peptide-1 (GLP-1) secretion. This action has been attributed to activation of the G protein-coupled bile acid receptor GPBAR1 (TGR5), although other potential bile acid sensors include the nuclear farnesoid receptor and the apical sodium-coupled bile acid transporter ASBT. The aim of this study was to identify pathways important for GLP-1 release and to determine whether bile acids target their receptors on GLP-1-secreting L-cells from the apical or basolateral compartment. Using transgenic mice expressing fluorescent sensors specifically in L-cells, we observed that taurodeoxycholate (TDCA) and taurolithocholate (TLCA) increased intracellular cAMP and Ca(2+). In primary intestinal cultures, TDCA was a more potent GLP-1 secretagogue than taurocholate (TCA) and TLCA, correlating with a stronger Ca(2+) response to TDCA. Using small-volume Ussing chambers optimized for measuring GLP-1 secretion, we found that both a GPBAR1 agonist and TDCA stimulated GLP-1 release better when applied from the basolateral than from the luminal direction and that luminal TDCA was ineffective when intestinal tissue was pretreated with an ASBT inhibitor. ASBT inhibition had no significant effect in nonpolarized primary cultures. Studies in the perfused rat gut confirmed that vascularly administered TDCA was more effective than luminal TDCA. Intestinal primary cultures and Ussing chamber-mounted tissues from GPBAR1-knockout mice did not secrete GLP-1 in response to either TLCA or TDCA. We conclude that the action of bile acids on GLP-1 secretion is predominantly mediated by GPBAR1 located on the basolateral L-cell membrane, suggesting that stimulation of gut hormone secretion may include postabsorptive mechanisms.
Collapse
|
1014
|
Palau-Rodriguez M, Tulipani S, Isabel Queipo-Ortuño M, Urpi-Sarda M, Tinahones FJ, Andres-Lacueva C. Metabolomic insights into the intricate gut microbial-host interaction in the development of obesity and type 2 diabetes. Front Microbiol 2015; 6:1151. [PMID: 26579078 PMCID: PMC4621279 DOI: 10.3389/fmicb.2015.01151] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 10/05/2015] [Indexed: 12/31/2022] Open
Abstract
Gut microbiota has recently been proposed as a crucial environmental factor in the development of metabolic diseases such as obesity and type 2 diabetes, mainly due to its contribution in the modulation of several processes including host energy metabolism, gut epithelial permeability, gut peptide hormone secretion, and host inflammatory state. Since the symbiotic interaction between the gut microbiota and the host is essentially reflected in specific metabolic signatures, much expectation is placed on the application of metabolomic approaches to unveil the key mechanisms linking the gut microbiota composition and activity with disease development. The present review aims to summarize the gut microbial-host co-metabolites identified so far by targeted and untargeted metabolomic studies in humans, in association with impaired glucose homeostasis and/or obesity. An alteration of the co-metabolism of bile acids, branched fatty acids, choline, vitamins (i.e., niacin), purines, and phenolic compounds has been associated so far with the obese or diabese phenotype, in respect to healthy controls. Furthermore, anti-diabetic treatments such as metformin and sulfonylurea have been observed to modulate the gut microbiota or at least their metabolic profiles, thereby potentially affecting insulin resistance through indirect mechanisms still unknown. Despite the scarcity of the metabolomic studies currently available on the microbial-host crosstalk, the data-driven results largely confirmed findings independently obtained from in vitro and animal model studies, putting forward the mechanisms underlying the implication of a dysfunctional gut microbiota in the development of metabolic disorders.
Collapse
Affiliation(s)
- Magali Palau-Rodriguez
- Biomarkers and Nutrimetabolomic Lab., Nutrition and Food Science Department, XaRTA, INSA, Campus Torribera, Pharmacy Faculty, University of Barcelona Barcelona, Spain
| | - Sara Tulipani
- Biomarkers and Nutrimetabolomic Lab., Nutrition and Food Science Department, XaRTA, INSA, Campus Torribera, Pharmacy Faculty, University of Barcelona Barcelona, Spain ; Biomedical Research Institute (IBIMA), Service of Endocrinology and Nutrition, Malaga Hospital Complex (Virgen de la Victoria), University of Malaga Malaga, Spain
| | - Maria Isabel Queipo-Ortuño
- Biomedical Research Institute (IBIMA), Service of Endocrinology and Nutrition, Malaga Hospital Complex (Virgen de la Victoria), University of Malaga Malaga, Spain ; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III (ISCIII) Madrid, Spain
| | - Mireia Urpi-Sarda
- Biomarkers and Nutrimetabolomic Lab., Nutrition and Food Science Department, XaRTA, INSA, Campus Torribera, Pharmacy Faculty, University of Barcelona Barcelona, Spain
| | - Francisco J Tinahones
- Biomedical Research Institute (IBIMA), Service of Endocrinology and Nutrition, Malaga Hospital Complex (Virgen de la Victoria), University of Malaga Malaga, Spain ; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III (ISCIII) Madrid, Spain
| | - Cristina Andres-Lacueva
- Biomarkers and Nutrimetabolomic Lab., Nutrition and Food Science Department, XaRTA, INSA, Campus Torribera, Pharmacy Faculty, University of Barcelona Barcelona, Spain
| |
Collapse
|
1015
|
Baars A, Oosting A, Knol J, Garssen J, van Bergenhenegouwen J. The Gut Microbiota as a Therapeutic Target in IBD and Metabolic Disease: A Role for the Bile Acid Receptors FXR and TGR5. Microorganisms 2015; 3:641-66. [PMID: 27682110 PMCID: PMC5023267 DOI: 10.3390/microorganisms3040641] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 10/01/2015] [Indexed: 12/18/2022] Open
Abstract
The gut microbiota plays a crucial role in regulating many physiological systems of the host, including the metabolic and immune system. Disturbances in microbiota composition are increasingly correlated with disease; however, the underlying mechanisms are not well understood. Recent evidence suggests that changes in microbiota composition directly affect the metabolism of bile salts. Next to their role in digestion of dietary fats, bile salts function as signaling molecules for bile salt receptors such as Farnesoid X receptor (FXR) and G protein-coupled bile acid receptor (TGR5). Complementary to their role in metabolism, FXR and TGR5 are shown to play a role in intestinal homeostasis and immune regulation. This review presents an overview of evidence showing that changes in bile salt pool and composition due to changes in gut microbial composition contribute to the pathogenesis of inflammatory bowel disease and metabolic disease, possibly through altered activation of TGR5 and FXR. We further discuss how dietary interventions, such as pro- and synbiotics, may be used to treat metabolic disease and inflammatory bowel disease (IBD) through normalization of bile acid dysregulation directly or indirectly through normalization of the intestinal microbiota.
Collapse
Affiliation(s)
| | | | - Jan Knol
- Nutricia Research, 3584 CT, Utrecht, The Netherlands.
- Laboratory of Microbiology, Wageningen University, 6703 HB, Wageningen, The Netherlands.
| | - Johan Garssen
- Nutricia Research, 3584 CT, Utrecht, The Netherlands.
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG, Utrecht, The Netherlands.
| | - Jeroen van Bergenhenegouwen
- Nutricia Research, 3584 CT, Utrecht, The Netherlands.
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG, Utrecht, The Netherlands.
| |
Collapse
|
1016
|
Barlow GM, Yu A, Mathur R. Role of the Gut Microbiome in Obesity and Diabetes Mellitus. Nutr Clin Pract 2015; 30:787-97. [PMID: 26452391 DOI: 10.1177/0884533615609896] [Citation(s) in RCA: 175] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) and obesity represent two of the biggest global health challenges of this century and are associated with significant comorbidities and healthcare costs. Although multiple factors undoubtedly contribute to the development and progression of DM and obesity, research over the last decade has demonstrated that the microbes that colonize the human gut may play key contributory roles. Gut microbes are now known to codevelop with the human host and are strongly influenced by mode of birth and early diet and nutrition, as well as environmental and other factors including antibiotic exposure. Gut microbes contribute to human health through roles in polysaccharide breakdown, nutrient absorption, inflammatory responses, gut permeability, and bile acid modification. Numerous studies have suggested that disruptions in the relative proportions of gut microbial populations may contribute to weight gain and insulin resistance, including alterations in Gammaproteobacteria and Verrucomicrobia and the ratios of Firmicutes to Bacteroidetes in weight gain and possible alterations in butyrate-producing bacteria such as Faecalibacterium prausnitzii in DM. In addition, it has been shown that the methanogenic Archaea may contribute to altered metabolism and weight gain in the host. However, the majority of studies are performed with stool or colonic samples and may not be representative of the metabolically active small intestine. Studies predominantly in rodent models are beginning to elucidate the mechanisms by which gut microbes contribute to DM and obesity, but much remains to be learned before we can begin to approach targeted treatments.
Collapse
Affiliation(s)
- Gillian M Barlow
- GI Motility Program, Cedars-Sinai Medical Center, Los Angeles, California
| | - Allen Yu
- GI Motility Program, Cedars-Sinai Medical Center, Los Angeles, California
| | - Ruchi Mathur
- Division of Endocrine Diabetes and Metabolism, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|
1017
|
Abstract
The enteroendocrine system orchestrates how the body responds to the ingestion of foods, employing a diversity of hormones to fine-tune a wide range of physiological responses both within and outside the gut. Recent interest in gut hormones has surged with the realization that they modulate glucose tolerance and food intake through a variety of mechanisms, and such hormones are therefore excellent therapeutic candidates for the treatment of diabetes and obesity. Characterizing the roles and functions of different enteroendocrine cells is an essential step in understanding the physiology, pathophysiology, and therapeutics of the gut-brain-pancreas axis.
Collapse
Affiliation(s)
- Fiona M Gribble
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, and Addenbrooke's Hospital, University of Cambridge, Cambridge CB2 0QQ, United Kingdom; ,
| | - Frank Reimann
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, and Addenbrooke's Hospital, University of Cambridge, Cambridge CB2 0QQ, United Kingdom; ,
| |
Collapse
|
1018
|
Klaassen CD, Cui JY. Review: Mechanisms of How the Intestinal Microbiota Alters the Effects of Drugs and Bile Acids. Drug Metab Dispos 2015; 43:1505-21. [PMID: 26261286 PMCID: PMC4576672 DOI: 10.1124/dmd.115.065698] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 08/05/2015] [Indexed: 12/27/2022] Open
Abstract
Information on the intestinal microbiota has increased exponentially this century because of technical advancements in genomics and metabolomics. Although information on the synthesis of bile acids by the liver and their transformation to secondary bile acids by the intestinal microbiota was the first example of the importance of the intestinal microbiota in biotransforming chemicals, this review will discuss numerous examples of the mechanisms by which the intestinal microbiota alters the pharmacology and toxicology of drugs and other chemicals. More specifically, the altered pharmacology and toxicology of salicylazosulfapridine, digoxin, l-dopa, acetaminophen, caffeic acid, phosphatidyl choline, carnitine, sorivudine, irinotecan, nonsteroidal anti-inflammatory drugs, heterocyclic amines, melamine, nitrazepam, and lovastatin will be reviewed. In addition, recent data that the intestinal microbiota alters drug metabolism of the host, especially Cyp3a, as well as the significance and potential mechanisms of this phenomenon are summarized. The review will conclude with an update of bile acid research, emphasizing the bile acid receptors (FXR and TGR5) that regulate not only bile acid synthesis and transport but also energy metabolism. Recent data indicate that by altering the intestinal microbiota, either by diet or drugs, one may be able to minimize the adverse effects of the Western diet by altering the composition of bile acids in the intestine that are agonists or antagonists of FXR and TGR5. Therefore, it may be possible to consider the intestinal microbiota as another drug target.
Collapse
Affiliation(s)
- Curtis D Klaassen
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| |
Collapse
|
1019
|
Singh TP, Vangaveti VN, Malabu UH. Dipeptidyl peptidase-4 inhibitors and their potential role in the management of atherosclerosis--A review. Diabetes Metab Syndr 2015; 9:223-229. [PMID: 26067782 DOI: 10.1016/j.dsx.2015.04.005] [Citation(s) in RCA: 13] [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] [Indexed: 01/22/2023]
Abstract
BACKGROUND Dipeptidyl peptidase-4 (DPP-4) inhibitors are a relatively new class of anti-diabetic drugs that have therapeutic potential in the management of atherosclerosis. Of the numerous DPP-4 inhibitors in clinical practice no studies have been conducted to compare their anti-atherosclerotic effects despite growing evidence of their usefulness in the high risk population. OBJECTIVE The aim of the study was to review the anti-athero-thrombotic effects of DPP-4 inhibitors in atherosclerotic diseases. METHOD A literature search was conducted on MEDLINE and the COCHRANE Library using the terms "DPP-4 Inhibitors", "atherosclerosis", "GLP", "inflammation", "cytokines", "stroke", "ischaemic heart disease", "hypertension" and "peripheral vascular disease". A mathematical model devised by us was used to derive and compare the anti-inflammatory effects of the DPP-4 inhibitors using protective score [PS]. Data was analysed for alogliptin, linagliptin, saxagliptin, sitagliptin, and vildagliptin. RESULTS Sixty two published studies collected in the search were assessed for relevance to this study. Sitagliptin had the highest PS [n=5] while linagliptin and saxagliptin yielded the lowest PS [n=1]. This comparison and scoring system was limited to the data collected; which did not investigate for all athero-thrombotic factors selected in this study. CONCLUSION The findings suggest potential usefulness of DPP-4 in atherosclerotic diseases. It also reflected a possible superiority of sitagliptin over the other DPP-4 inhibitors in the management of atherosclerosis. Further investigations are required to establish specific inflammatory cytokines influenced by the DPP-4 inhibitors and to elucidate their clinical application in athero-thrombotic disease.
Collapse
Affiliation(s)
- Tejas P Singh
- Translational Research on Endocrinology and Diabetes [TREAD], School of Medicine and Dentistry, James Cook University Townsville, Australia
| | - Venkat N Vangaveti
- Translational Research on Endocrinology and Diabetes [TREAD], School of Medicine and Dentistry, James Cook University Townsville, Australia
| | - Usman H Malabu
- Translational Research on Endocrinology and Diabetes [TREAD], School of Medicine and Dentistry, James Cook University Townsville, Australia.
| |
Collapse
|
1020
|
Selwyn FP, Csanaky IL, Zhang Y, Klaassen CD. Importance of Large Intestine in Regulating Bile Acids and Glucagon-Like Peptide-1 in Germ-Free Mice. Drug Metab Dispos 2015; 43:1544-56. [PMID: 26199423 PMCID: PMC4576674 DOI: 10.1124/dmd.115.065276] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 07/20/2015] [Indexed: 12/25/2022] Open
Abstract
It is known that 1) elevated serum bile acids (BAs) are associated with decreased body weight, 2) elevated glucagon-like peptide-1 (GLP-1) levels can decrease body weight, and 3) germ-free (GF) mice are resistant to diet-induced obesity. The purpose of this study was to test the hypothesis that a lack of intestinal microbiota results in more BAs in the body, resulting in increased BA-mediated transmembrane G protein-coupled receptor 5 (TGR5) signaling and increased serum GLP-1 as a mechanism of resistance of GF mice to diet-induced obesity. GF mice had 2- to 4-fold increased total BAs in the serum, liver, bile, and ileum. Fecal excretion of BAs was 63% less in GF mice. GF mice had decreased secondary BAs and increased taurine-conjugated BAs, as anticipated. Surprisingly, there was an increase in non-12α-OH BAs, namely, β-muricholic acid, ursodeoxycholic acid (UDCA), and their taurine conjugates, in GF mice. Further, in vitro experiments confirmed that UDCA is a primary BA in mice. There were minimal changes in the mRNA of farnesoid X receptor target genes in the ileum (Fibroblast growth factor 15, small heterodimer protein, and ileal bile acid-binding protein), in the liver (small heterodimer protein, liver receptor homolog-1, and cytochrome P450 7a1), and BA transporters (apical sodium dependent bile acid transporter, organic solute transporter α, and organic solute transporter β) in the ileum of GF mice. Surprisingly, there were marked increases in BA transporters in the large intestine. Increased GLP-1 levels and gallbladder size were observed in GF mice, suggesting activation of TGR5 signaling. In summary, the GF condition results in increased expression of BA transporters in the colon, resulting in 1) an increase in total BA concentrations in tissues, 2) a change in BA composition to favor an increase in non-12α-OH BAs, and 3) activation of TGR5 signaling with increased gallbladder size and GLP-1.
Collapse
Affiliation(s)
- Felcy Pavithra Selwyn
- Departments of Pharmacology, Toxicology, and Therapeutics (F.P.S., Y.Z.) and Internal Medicine (I.L.C., C.D.K.), University of Kansas Medical Center, Kansas City, Kansas
| | - Iván L Csanaky
- Departments of Pharmacology, Toxicology, and Therapeutics (F.P.S., Y.Z.) and Internal Medicine (I.L.C., C.D.K.), University of Kansas Medical Center, Kansas City, Kansas
| | - Youcai Zhang
- Departments of Pharmacology, Toxicology, and Therapeutics (F.P.S., Y.Z.) and Internal Medicine (I.L.C., C.D.K.), University of Kansas Medical Center, Kansas City, Kansas
| | - Curtis D Klaassen
- Departments of Pharmacology, Toxicology, and Therapeutics (F.P.S., Y.Z.) and Internal Medicine (I.L.C., C.D.K.), University of Kansas Medical Center, Kansas City, Kansas.
| |
Collapse
|
1021
|
Delzenne NM, Cani PD, Everard A, Neyrinck AM, Bindels LB. Gut microorganisms as promising targets for the management of type 2 diabetes. Diabetologia 2015. [PMID: 26224102 DOI: 10.1007/s00125-015-3712-7] [Citation(s) in RCA: 196] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Each human intestine harbours not only hundreds of trillions of bacteria but also bacteriophage particles, viruses, fungi and archaea, which constitute a complex and dynamic ecosystem referred to as the gut microbiota. An increasing number of data obtained during the last 10 years have indicated changes in gut bacterial composition or function in type 2 diabetic patients. Analysis of this 'dysbiosis' enables the detection of alterations in specific bacteria, clusters of bacteria or bacterial functions associated with the occurrence or evolution of type 2 diabetes; these bacteria are predominantly involved in the control of inflammation and energy homeostasis. Our review focuses on two key questions: does gut dysbiosis truly play a role in the occurrence of type 2 diabetes, and will recent discoveries linking the gut microbiota to host health be helpful for the development of novel therapeutic approaches for type 2 diabetes? Here we review how pharmacological, surgical and nutritional interventions for type 2 diabetic patients may impact the gut microbiota. Experimental studies in animals are identifying which bacterial metabolites and components act on host immune homeostasis and glucose metabolism, primarily by targeting intestinal cells involved in endocrine and gut barrier functions. We discuss novel approaches (e.g. probiotics, prebiotics and faecal transfer) and the need for research and adequate intervention studies to evaluate the feasibility and relevance of these new therapies for the management of type 2 diabetes.
Collapse
Affiliation(s)
- Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Avenue E. Mounier, 73, B1.73.11, 1200, Brussels, Belgium.
| | - Patrice D Cani
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Avenue E. Mounier, 73, B1.73.11, 1200, Brussels, Belgium
- Walloon Excellence in Life sciences and BIOtechnology (WELBIO), Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Amandine Everard
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Avenue E. Mounier, 73, B1.73.11, 1200, Brussels, Belgium
| | - Audrey M Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Avenue E. Mounier, 73, B1.73.11, 1200, Brussels, Belgium
| | - Laure B Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Avenue E. Mounier, 73, B1.73.11, 1200, Brussels, Belgium
| |
Collapse
|
1022
|
Wang XX, Edelstein MH, Gafter U, Qiu L, Luo Y, Dobrinskikh E, Lucia S, Adorini L, D'Agati VD, Levi J, Rosenberg A, Kopp JB, Gius DR, Saleem MA, Levi M. G Protein-Coupled Bile Acid Receptor TGR5 Activation Inhibits Kidney Disease in Obesity and Diabetes. J Am Soc Nephrol 2015; 27:1362-78. [PMID: 26424786 DOI: 10.1681/asn.2014121271] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 08/12/2015] [Indexed: 12/31/2022] Open
Abstract
Obesity and diabetes mellitus are the leading causes of renal disease. In this study, we determined the regulation and role of the G protein-coupled bile acid receptor TGR5, previously shown to be regulated by high glucose and/or fatty acids, in obesity-related glomerulopathy (ORG) and diabetic nephropathy (DN). Treatment of diabetic db/db mice with the selective TGR5 agonist INT-777 decreased proteinuria, podocyte injury, mesangial expansion, fibrosis, and CD68 macrophage infiltration in the kidney. INT-777 also induced renal expression of master regulators of mitochondrial biogenesis, inhibitors of oxidative stress, and inducers of fatty acid β-oxidation, including sirtuin 1 (SIRT1), sirtuin 3 (SIRT3), and Nrf-1. Increased activity of SIRT3 was evidenced by normalization of the increased acetylation of mitochondrial superoxide dismutase 2 (SOD2) and isocitrate dehydrogenase 2 (IDH2) observed in untreated db/db mice. Accordingly, INT-777 decreased mitochondrial H2O2 generation and increased the activity of SOD2, which associated with decreased urinary levels of H2O2 and thiobarbituric acid reactive substances. Furthermore, INT-777 decreased renal lipid accumulation. INT-777 also prevented kidney disease in mice with diet-induced obesity. In human podocytes cultured with high glucose, INT-777 induced mitochondrial biogenesis, decreased oxidative stress, and increased fatty acid β-oxidation. Compared with normal kidney biopsy specimens, kidney specimens from patients with established ORG or DN expressed significantly less TGR5 mRNA, and levels inversely correlated with disease progression. Our results indicate that TGR5 activation induces mitochondrial biogenesis and prevents renal oxidative stress and lipid accumulation, establishing a role for TGR5 in inhibiting kidney disease in obesity and diabetes.
Collapse
Affiliation(s)
- Xiaoxin X Wang
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado;
| | - Michal Herman Edelstein
- Rabin Medical Center, Department of Nephrology and Hypertension; Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Israel
| | - Uzi Gafter
- Rabin Medical Center, Department of Nephrology and Hypertension; Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Israel
| | - Liru Qiu
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Yuhuan Luo
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Evgenia Dobrinskikh
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Scott Lucia
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | | | - Vivette D D'Agati
- Department of Pathology, Columbia University, College of Physicians and Surgeons, New York, New York
| | - Jonathan Levi
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Avi Rosenberg
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Jeffrey B Kopp
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - David R Gius
- Northwestern University Feinberg School of Medicine, Chicago, Illinois; and
| | - Moin A Saleem
- University of Bristol, Southmead Hospital, Bristol, United Kingdom
| | - Moshe Levi
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado;
| |
Collapse
|
1023
|
Gertzen CGW, Spomer L, Smits SHJ, Häussinger D, Keitel V, Gohlke H. Mutational mapping of the transmembrane binding site of the G-protein coupled receptor TGR5 and binding mode prediction of TGR5 agonists. Eur J Med Chem 2015; 104:57-72. [PMID: 26435512 DOI: 10.1016/j.ejmech.2015.09.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Revised: 09/06/2015] [Accepted: 09/15/2015] [Indexed: 12/31/2022]
Abstract
TGR5 (Gpbar-1, M-Bar) is a class A G-protein coupled bile acid-sensing receptor predominately expressed in brain, liver and gastrointestinal tract, and a promising drug target for the treatment of metabolic disorders. Due to the lack of a crystal structure of TGR5, the development of TGR5 agonists has been guided by ligand-based approaches so far. Three binding mode models of bile acid derivatives have been presented recently. However, they differ from one another in terms of overall orientation or with respect to the location and interactions of the cholane scaffold, or cannot explain all results from mutagenesis experiments. Here, we present an extended binding mode model based on an iterative and integrated computational and biological approach. An alignment of 68 TGR5 agonists based on this binding mode leads to a significant and good structure-based 3D QSAR model, which constitutes the most comprehensive structure-based 3D-QSAR study of TGR5 agonists undertaken so far and suggests that the binding mode model is a close representation of the "true" binding mode. The binding mode model is further substantiated in that effects predicted for eight mutations in the binding site agree with experimental analyses on the impact of these TGR5 variants on receptor activity. In the binding mode, the hydrophobic cholane scaffold of taurolithocholate orients towards the interior of the orthosteric binding site such that rings A and B are in contact with TM5 and TM6, the taurine side chain orients towards the extracellular opening of the binding site and forms a salt bridge with R79(EL1), and the 3-hydroxyl group forms hydrogen bonds with E169(5.44) and Y240(6.51). The binding mode thus differs in important aspects from the ones recently presented. These results are highly relevant for the development of novel, more potent agonists of TGR5 and should be a valuable starting point for the development of TGR5 antagonists, which could show antiproliferative effects in tumor cells.
Collapse
Affiliation(s)
- Christoph G W Gertzen
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich Heine University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Lina Spomer
- Clinic for Gastroenterology, Hepatology, and Infectious Diseases, Heinrich Heine University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Sander H J Smits
- Institute for Biochemistry, Heinrich Heine University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Dieter Häussinger
- Clinic for Gastroenterology, Hepatology, and Infectious Diseases, Heinrich Heine University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Verena Keitel
- Clinic for Gastroenterology, Hepatology, and Infectious Diseases, Heinrich Heine University, Universitätsstr. 1, 40225 Düsseldorf, Germany.
| | - Holger Gohlke
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich Heine University, Universitätsstr. 1, 40225 Düsseldorf, Germany.
| |
Collapse
|
1024
|
Mooranian A, Negrulj R, Arfuso F, Al-Salami H. Multicompartmental, multilayered probucol microcapsules for diabetes mellitus: Formulation characterization and effects on production of insulin and inflammation in a pancreatic β-cell line. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2015; 44:1642-53. [DOI: 10.3109/21691401.2015.1069299] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
1025
|
Brown adipose tissue: a potential target in the fight against obesity and the metabolic syndrome. Clin Sci (Lond) 2015; 129:933-49. [DOI: 10.1042/cs20150339] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BAT (brown adipose tissue) is the main site of thermogenesis in mammals. It is essential to ensure thermoregulation in newborns. It is also found in (some) adult humans. Its capacity to oxidize fatty acids and glucose without ATP production contributes to energy expenditure and glucose homoeostasis. Brown fat activation has thus emerged as an attractive therapeutic target for the treatment of obesity and the metabolic syndrome. In the present review, we integrate the recent advances on the metabolic role of BAT and its relation with other tissues as well as its potential contribution to fighting obesity and the metabolic syndrome.
Collapse
|
1026
|
Stinkens R, Goossens GH, Jocken JWE, Blaak EE. Targeting fatty acid metabolism to improve glucose metabolism. Obes Rev 2015; 16:715-57. [PMID: 26179344 DOI: 10.1111/obr.12298] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 04/23/2015] [Accepted: 05/10/2015] [Indexed: 12/15/2022]
Abstract
Disturbances in fatty acid metabolism in adipose tissue, liver, skeletal muscle, gut and pancreas play an important role in the development of insulin resistance, impaired glucose metabolism and type 2 diabetes mellitus. Alterations in diet composition may contribute to prevent and/or reverse these disturbances through modulation of fatty acid metabolism. Besides an increased fat mass, adipose tissue dysfunction, characterized by an altered capacity to store lipids and an altered secretion of adipokines, may result in lipid overflow, systemic inflammation and excessive lipid accumulation in non-adipose tissues like liver, skeletal muscle and the pancreas. These impairments together promote the development of impaired glucose metabolism, insulin resistance and type 2 diabetes mellitus. Furthermore, intrinsic functional impairments in either of these organs may contribute to lipotoxicity and insulin resistance. The present review provides an overview of fatty acid metabolism-related pathways in adipose tissue, liver, skeletal muscle, pancreas and gut, which can be targeted by diet or food components, thereby improving glucose metabolism.
Collapse
Affiliation(s)
- R Stinkens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - G H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - J W E Jocken
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - E E Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| |
Collapse
|
1027
|
Δ(5)-Cholenoyl-amino acids as selective and orally available antagonists of the Eph-ephrin system. Eur J Med Chem 2015; 103:312-24. [PMID: 26363867 DOI: 10.1016/j.ejmech.2015.08.048] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 08/24/2015] [Accepted: 08/25/2015] [Indexed: 01/22/2023]
Abstract
The Eph receptor-ephrin system is an emerging target for the development of novel anti-angiogenic therapies. Research programs aimed at developing small-molecule antagonists of the Eph receptors are still in their initial stage as available compounds suffer from pharmacological drawbacks, limiting their application in vitro and in vivo. In the present work, we report the design, synthesis and evaluation of structure-activity relationships of a class of Δ(5)-cholenoyl-amino acid conjugates as Eph-ephrin antagonists. As a major achievement of our exploration, we identified N-(3β-hydroxy-Δ(5)-cholen-24-oyl)-L-tryptophan (UniPR1331) as the first small molecule antagonist of the Eph-ephrin system effective as an anti-angiogenic agent in endothelial cells, bioavailable in mice by the oral route and devoid of biological activity on G protein-coupled and nuclear receptors targeted by bile acid derivatives.
Collapse
|
1028
|
Abstract
Bile acids are well known for their effects on cholesterol homeostasis and lipid digestion. Since the discovery of bile acid receptors, of which there are farnesoid X receptor (FXR), a nuclear receptor, and the plasma membrane G-protein receptor, as well as Takeda G-protein coupled receptor clone 5, further roles have been elucidated for bile acids including glucose and lipid metabolism as well as inflammation. Additionally, treatment with bile acid receptor agonists has shown a decrease in the amount of atherosclerosis plaque formation and decreased portal vascular resistance and portal hypotension in animal models. Furthermore, rodent models have demonstrated antifibrotic activity using bile acid receptor agonists. Early human data using a FXR agonist, obeticholic acid, have shown promising results with improvement of histological activity and even a reduction of fibrosis. Human studies are ongoing and will provide further information on bile acid receptor agonist therapies. Thus, bile acids and their derivatives have the potential for management of liver diseases and potentially other disease states including diabetes and the metabolic syndrome.
Collapse
|
1029
|
Briere DA, Ruan X, Cheng CC, Siesky AM, Fitch TE, Dominguez C, Sanfeliciano SG, Montero C, Suen CS, Xu Y, Coskun T, Michael MD. Novel Small Molecule Agonist of TGR5 Possesses Anti-Diabetic Effects but Causes Gallbladder Filling in Mice. PLoS One 2015; 10:e0136873. [PMID: 26312995 PMCID: PMC4551797 DOI: 10.1371/journal.pone.0136873] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 08/09/2015] [Indexed: 12/25/2022] Open
Abstract
Activation of TGR5 via bile acids or bile acid analogs leads to the release of glucagon-like peptide-1 (GLP-1) from intestine, increases energy expenditure in brown adipose tissue, and increases gallbladder filling with bile. Here, we present compound 18, a non-bile acid agonist of TGR5 that demonstrates robust GLP-1 secretion in a mouse enteroendocrine cell line yet weak GLP-1 secretion in a human enteroendocrine cell line. Acute administration of compound 18 to mice increased GLP-1 and peptide YY (PYY) secretion, leading to a lowering of the glucose excursion in an oral glucose tolerance test (OGTT), while chronic administration led to weight loss. In addition, compound 18 showed a dose-dependent increase in gallbladder filling. Lastly, compound 18 failed to show similar pharmacological effects on GLP-1, PYY, and gallbladder filling in Tgr5 knockout mice. Together, these results demonstrate that compound 18 is a mouse-selective TGR5 agonist that induces GLP-1 and PYY secretion, and lowers the glucose excursion in an OGTT, but only at doses that simultaneously induce gallbladder filling. Overall, these data highlight the benefits and potential risks of using TGR5 agonists to treat diabetes and metabolic diseases.
Collapse
Affiliation(s)
- Daniel A. Briere
- Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, United States of America
| | - Xiaoping Ruan
- Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, United States of America
| | - Christine C. Cheng
- Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, United States of America
| | - Angela M. Siesky
- Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, United States of America
| | - Thomas E. Fitch
- Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, United States of America
| | - Carmen Dominguez
- Centro de Investigación, Eli Lilly and Company, Alcobendas, Spain
| | | | - Carlos Montero
- Centro de Investigación, Eli Lilly and Company, Alcobendas, Spain
| | - Chen S. Suen
- Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, United States of America
| | - Yanping Xu
- Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, United States of America
| | - Tamer Coskun
- Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, United States of America
| | - M. Dodson Michael
- Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana, United States of America
- * E-mail:
| |
Collapse
|
1030
|
Sawitza I, Kordes C, Götze S, Herebian D, Häussinger D. Bile acids induce hepatic differentiation of mesenchymal stem cells. Sci Rep 2015; 5:13320. [PMID: 26304833 PMCID: PMC4548444 DOI: 10.1038/srep13320] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 07/23/2015] [Indexed: 12/25/2022] Open
Abstract
Mesenchymal stem cells (MSC) have the potential to differentiate into multiple cell lineages and their therapeutic potential has become obvious. In the liver, MSC are represented by stellate cells which have the potential to differentiate into hepatocytes after stimulation with growth factors. Since bile acids can promote liver regeneration, their influence on liver-resident and bone marrow-derived MSC was investigated. Physiological concentrations of bile acids such as tauroursodeoxycholic acid were able to initiate hepatic differentiation of MSC via the farnesoid X receptor and transmembrane G-protein-coupled bile acid receptor 5 as investigated with knockout mice. Notch, hedgehog, transforming growth factor-β/bone morphogenic protein family and non-canonical Wnt signalling were also essential for bile acid-mediated differentiation, whereas β-catenin-dependent Wnt signalling was able to attenuate this process. Our findings reveal bile acid-mediated signalling as an alternative way to induce hepatic differentiaion of stem cells and highlight bile acids as important signalling molecules during liver regeneration.
Collapse
Affiliation(s)
- Iris Sawitza
- Clinic of Gastroenterology, Hepatology and Infectious Diseases
| | - Claus Kordes
- Clinic of Gastroenterology, Hepatology and Infectious Diseases
| | - Silke Götze
- Clinic of Gastroenterology, Hepatology and Infectious Diseases
| | - Diran Herebian
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Heinrich Heine University, Moorenstraße 5, 40225 Düsseldorf, Germany
| | | |
Collapse
|
1031
|
López-García S, Castañeda-Sanchez JI, Jiménez-Arellanes A, Domínguez-López L, Castro-Mussot ME, Hernández-Sanchéz J, Luna-Herrera J. Macrophage Activation by Ursolic and Oleanolic Acids during Mycobacterial Infection. Molecules 2015; 20:14348-64. [PMID: 26287131 PMCID: PMC6332297 DOI: 10.3390/molecules200814348] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Revised: 07/30/2015] [Accepted: 07/31/2015] [Indexed: 12/31/2022] Open
Abstract
Oleanolic (OA) and ursolic acids (UA) are triterpenes that are abundant in vegetables, fruits and medicinal plants. They have been described as active moieties in medicinal plants used for the treatment of tuberculosis. In this study, we analyzed the effects of these triterpenes on macrophages infected in vitro with Mycobacterium tuberculosis (MTB). We evaluated production of nitric oxide (NO), reactive oxygen species (ROS), and cytokines (TNF-α and TGF-β) as well as expression of cell membrane receptors (TGR5 and CD36) in MTB-infected macrophages following treatment with OA and UA. Triterpenes caused reduced MTB growth in macrophages, stimulated production of NO and ROS in the early phase, stimulated TNF-α, suppressed TGF-β and caused over-expression of CD36 and TGR5 receptors. Thus, our data suggest immunomodulatory properties of OA and UA on MTB infected macrophages. In conclusion, antimycobacterial effects induced by these triterpenes may be attributable to the conversion of macrophages from stage M2 (alternatively activated) to M1 (classically activated).
Collapse
Affiliation(s)
- Sonia López-García
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, IPN, Prolongación de Carpio y Plan de Ayala S/N, 11340 México City, Mexico; E-Mails: (S.L.-G.); (L.D.-L.); (M.E.C.-M.)
- Departamento de Genética y Biología Molecular, CINVESTAV, Avenida Instituto Politécnico Nacional Número 2508, 07360 México City, Mexico; E-Mail:
| | - Jorge Ismael Castañeda-Sanchez
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana, Unidad Xochimilco, Calzada del Hueso Número 1100, 04960 México City, Mexico; E-Mail:
| | - Adelina Jiménez-Arellanes
- Centro Médico Nacional Siglo XXI, IMSS, Unidad de Investigación Médica en Farmacología, Avenida Cuauhtémoc Número 330, 06725 México City, Mexico; E-Mail:
| | - Lilia Domínguez-López
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, IPN, Prolongación de Carpio y Plan de Ayala S/N, 11340 México City, Mexico; E-Mails: (S.L.-G.); (L.D.-L.); (M.E.C.-M.)
| | - Maria Eugenia Castro-Mussot
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, IPN, Prolongación de Carpio y Plan de Ayala S/N, 11340 México City, Mexico; E-Mails: (S.L.-G.); (L.D.-L.); (M.E.C.-M.)
| | - Javier Hernández-Sanchéz
- Departamento de Genética y Biología Molecular, CINVESTAV, Avenida Instituto Politécnico Nacional Número 2508, 07360 México City, Mexico; E-Mail:
| | - Julieta Luna-Herrera
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, IPN, Prolongación de Carpio y Plan de Ayala S/N, 11340 México City, Mexico; E-Mails: (S.L.-G.); (L.D.-L.); (M.E.C.-M.)
| |
Collapse
|
1032
|
Abstract
PURPOSE OF REVIEW To summarize recent epidemiological, preclinical and clinical studies on the effects of Roux-en-Y-gastric bypass (RYGBP) surgery on cardiovascular risk factors and the underlying mechanisms. RECENT FINDINGS Although RYGBP has mechanical effects on the gastrointestinal tract, the reduced gastric pouch and intestinal calorie absorption cannot fully explain the metabolic improvements. SUMMARY Obesity predisposes to cardiovascular risk factors such as dyslipidemia, type 2 diabetes, nonalcoholic fatty liver disease and hypertension. In contrast to the limited success of pharmacological and lifestyle interventions, RYGBP induces sustained weight loss, metabolic improvements and decreases morbidity/mortality. In line, RYGBP reduces cardiovascular risk factors. Although the mechanisms are not entirely understood, RYGBP induces complex changes in the gut affecting other organs through endocrine and metabolic signals from the intestine to all key metabolic organs, which can link RYGBP and decreased cardiovascular risk. Here, we discuss the roles of changes in lipid absorption and metabolism, bile acid metabolism, gut hormones and the microbiote as potential mechanisms in the decreased cardiovascular risk and metabolic improvement after RYGBP.
Collapse
Affiliation(s)
- Anne Tailleux
- aEuropean Genomic Institute for Diabetes (EGID) bINSERM UMR1011 cUniv Lille 2 dInstitut Pasteur de Lille, Lille, France eBiomedical Sciences Research Center 'Alexander Fleming', Vari, Greece fINSERM UMR1190, Lille, France
| | | | | | | |
Collapse
|
1033
|
Org E, Mehrabian M, Lusis AJ. Unraveling the environmental and genetic interactions in atherosclerosis: Central role of the gut microbiota. Atherosclerosis 2015; 241:387-99. [PMID: 26071662 PMCID: PMC4510029 DOI: 10.1016/j.atherosclerosis.2015.05.035] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 05/27/2015] [Accepted: 05/27/2015] [Indexed: 02/06/2023]
Abstract
Recent studies have convincingly linked gut microbiota to traits relevant to atherosclerosis, such as insulin resistance, dyslipidemia and inflammation, and have revealed novel disease pathways involving microbe-derived metabolites. These results have important implications for understanding how environmental and genetic factors act together to influence cardiovascular disease (CVD) risk. Thus, dietary constituents are not only absorbed and metabolized by the host but they also perturb the gut microbiota, which in turn influence host metabolism and inflammation. It also appears that host genetics helps to shape the gut microbiota community. Here, we discuss challenges in understanding these interactions and the role they play in CVD.
Collapse
Affiliation(s)
- Elin Org
- Departments of Medicine, Microbiology and Human Genetics, University of California, Los Angeles, CA, USA
| | - Margarete Mehrabian
- Departments of Medicine, Microbiology and Human Genetics, University of California, Los Angeles, CA, USA
| | - Aldons J Lusis
- Departments of Medicine, Microbiology and Human Genetics, University of California, Los Angeles, CA, USA.
| |
Collapse
|
1034
|
Horiba T, Katsukawa M, Mita M, Sato R. Dietary obacunone supplementation stimulates muscle hypertrophy, and suppresses hyperglycemia and obesity through the TGR5 and PPARγ pathway. Biochem Biophys Res Commun 2015; 463:846-52. [DOI: 10.1016/j.bbrc.2015.06.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 06/03/2015] [Indexed: 01/22/2023]
|
1035
|
Zheng C, Zhou W, Wang T, You P, Zhao Y, Yang Y, Wang X, Luo J, Chen Y, Liu M, Chen H. A Novel TGR5 Activator WB403 Promotes GLP-1 Secretion and Preserves Pancreatic β-Cells in Type 2 Diabetic Mice. PLoS One 2015; 10:e0134051. [PMID: 26208278 PMCID: PMC4514850 DOI: 10.1371/journal.pone.0134051] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Accepted: 07/04/2015] [Indexed: 12/31/2022] Open
Abstract
The G protein-coupled receptor TGR5 is a membrane receptor for bile acids. Its agonism increases energy expenditure and controls blood glucose through secretion of glucagon-like peptide-1 in enteroendocrine cells. In this study, we explored the therapeutic potential of WB403, a small compound activating TGR5 which was identified by combining TGR5 targeted luciferase assay and active GLP-1 assay, in treating type 2 diabetes. After confirmation of TGR5 and GLP-1 stimulating activities in various cell systems, WB403 was examined in oral glucose tolerance test, and tested on different mouse models of type 2 diabetes for glycemic control and pancreatic β-cell protection effect. As a result, WB403 exhibited a moderate TGR5 activation effect while promoting GLP-1 secretion efficiently. Interestingly, gallbladder filling effect, which was reported for some known TGR5 agonists, was not detected in this novel compound. In vivo results showed that WB403 significantly improved glucose tolerance and decreased fasting blood glucose, postprandial blood glucose and HbA1c in type 2 diabetic mice. Further analysis revealed that WB403 increased pancreatic β-cells and restored the normal distribution pattern of α-cell and β-cell in islets. These findings demonstrated that TGR5 activator WB403 effectively promoted GLP-1 release, improved hyperglycemia and preserved the mass and function of pancreatic β-cells, whereas it did not show a significant side effect on gallbladder. It may represent a promising approach for future type 2 diabetes mellitus drug development.
Collapse
Affiliation(s)
- Chunbing Zheng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Wenbo Zhou
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Tongtong Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Panpan You
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Yongliang Zhao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Yiqing Yang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Xin Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Jian Luo
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Yihua Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
- Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, Texas, United States of America
| | - Huaqing Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| |
Collapse
|
1036
|
Rudling M, Camilleri M, Graffner H, Holst JJ, Rikner L. Specific inhibition of bile acid transport alters plasma lipids and GLP-1. BMC Cardiovasc Disord 2015. [PMID: 26197999 PMCID: PMC4511433 DOI: 10.1186/s12872-015-0070-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Elobixibat is a minimally absorbed ileal bile acid (BA) transporter (IBAT) inhibitor in development against chronic constipation (CC) and constipation-predominant Irritable Bowel Syndrome (IBS-C). CC is associated with an increased risk for cardiovascular disease and type2 diabetes mellitus. The objectives of this study were to evaluate metabolic effects of elobixibat. Effects on plasma lipids and BA synthesis were evaluated utilizing a 4-week, placebo-controlled study in patients with dyslipidemia while changes of glucagon-like peptide-1 (GLP-1) by elobixibat was assayed in samples from a 14 day high-dose elobixibat study in patients with CC. METHODS Thirty-six dyslipidemic patients, 21 females, mean age 63 years, were randomized to 2.5 mg or 5 mg elobixibat or placebo once daily for four weeks. The primary endpoint was the change in low density lipoprotein (LDL) cholesterol. Secondary endpoints included other lipid parameters and serum 7α-hydroxy-4-cholesten-3-one (C4), a marker of BA (bile acid) synthesis. Another study, in 36 patients with CC treated with high dose elobixibat; 15 mg or 20 mg/day or placebo for 14 days, was evaluated for changes in GLP-1. RESULTS In the dyslipidemia study LDL cholesterol was reduced by 7.4 % (p = 0.044), and the LDL/HDL ratio was decreased by 18 % (p = 0.004). Serum C4 increased, indicating that BA synthesis was induced. No serious adverse events were recorded. In the CC study, GLP-1 increased significantly in both the 15 mg (20.7 ± 2.4 pmol/L; p = 0.03) and the 20 mg group (25.6 ± 4.9 pmol/L; p = 0.02). CONCLUSIONS Elobixibat reduces LDL cholesterol and LDL/HDL ratio and increase circulating peak GLP-1 levels, the latter in line with increased intestinal BA mediated responses in humans. TRIAL REGISTRATIONS ClinicalTrial.gov: NCT01069783 and NCT01038687 .
Collapse
Affiliation(s)
- Mats Rudling
- Department of Endocrinology, Metabolism and Diabetes, Metabolism Unit, Center for Endocrinology, Metabolism, and Diabetes, Karolinska Institute at Karolinska University Hospital Huddinge, S-141 86, Stockholm, Sweden. .,Department of Medicine, and Molecular Nutrition Unit, Karolinska Institute at Karolinska University Hospital Huddinge, S-141 86, Stockholm, Sweden. .,Department of Biosciences and Nutrition, Karolinska Institute at Karolinska University Hospital Huddinge, S-141 86, Stockholm, Sweden.
| | | | | | - Jens Juul Holst
- NNF Center for Basic Metabolic Research, the Panum Institute, University of Copenhagen, Copenhagen, Denmark.
| | | |
Collapse
|
1037
|
Bile diversion to the distal small intestine has comparable metabolic benefits to bariatric surgery. Nat Commun 2015. [PMID: 26197299 PMCID: PMC4518285 DOI: 10.1038/ncomms8715] [Citation(s) in RCA: 154] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Roux-en-Y gastric bypass (RYGB) is highly effective in reversing obesity and associated diabetes. Recent observations in humans suggest a contributing role of increased circulating bile acids in mediating such effects. Here we use a diet-induced obesity (DIO) mouse model and compare metabolic remission when bile flow is diverted through a gallbladder anastomosis to jejunum, ileum or duodenum (sham control). We find that only bile diversion to the ileum results in physiologic changes similar to RYGB, including sustained improvements in weight, glucose tolerance and hepatic steatosis despite differential effects on hepatic gene expression. Circulating free fatty acids and triglycerides decrease while bile acids increase, particularly conjugated tauro-β-muricholic acid, an FXR antagonist. Activity of the hepatic FXR/FGF15 signalling axis is reduced and associated with altered gut microbiota. Thus bile diversion, independent of surgical rearrangement of the gastrointestinal tract, imparts significant weight loss accompanied by improved glucose and lipid homeostasis that are hallmarks of RYGB.
Collapse
|
1038
|
Mace OJ, Tehan B, Marshall F. Pharmacology and physiology of gastrointestinal enteroendocrine cells. Pharmacol Res Perspect 2015. [PMID: 26213627 PMCID: PMC4506687 DOI: 10.1002/prp2.155] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Gastrointestinal (GI) polypeptides are secreted from enteroendocrine cells (EECs). Recent technical advances and the identification of endogenous and synthetic ligands have enabled exploration of the pharmacology and physiology of EECs. Enteroendocrine signaling pathways stimulating hormone secretion involve multiple nutrient transporters and G protein-coupled receptors (GPCRs), which are activated simultaneously under prevailing nutrient conditions in the intestine following a meal. The majority of studies investigate hormone secretion from EECs in response to single ligands and although the mechanisms behind how individual signaling pathways generate a hormonal output have been well characterized, our understanding of how these signaling pathways converge to generate a single hormone secretory response is still in its infancy. However, a picture is beginning to emerge of how nutrients and full, partial, or allosteric GPCR ligands differentially regulate the enteroendocrine system and its interaction with the enteric and central nervous system. So far, activation of multiple pathways underlies drug discovery efforts to harness the therapeutic potential of the enteroendocrine system to mimic the phenotypic changes observed in patients who have undergone Roux-en-Y gastric surgery. Typically obese patients exhibit ∼30% weight loss and greater than 80% of obese diabetics show remission of diabetes. Targeting combinations of enteroendocrine signaling pathways that work synergistically may manifest with significant, differentiated EEC secretory efficacy. Furthermore, allosteric modulators with their increased selectivity, self-limiting activity, and structural novelty may translate into more promising enteroendocrine drugs. Together with the potential to bias enteroendocrine GPCR signaling and/or to activate multiple divergent signaling pathways highlights the considerable range of therapeutic possibilities available. Here, we review the pharmacology and physiology of the EEC system.
Collapse
Affiliation(s)
- O J Mace
- Heptares Therapeutics Ltd BioPark, Broadwater Road, Welwyn Garden City, AL7 3AX, United Kingdom
| | - B Tehan
- Heptares Therapeutics Ltd BioPark, Broadwater Road, Welwyn Garden City, AL7 3AX, United Kingdom
| | - F Marshall
- Heptares Therapeutics Ltd BioPark, Broadwater Road, Welwyn Garden City, AL7 3AX, United Kingdom
| |
Collapse
|
1039
|
Traussnigg S, Kienbacher C, Halilbasic E, Rechling C, Kazemi-Shirazi L, Hofer H, Munda P, Trauner M. Challenges and Management of Liver Cirrhosis: Practical Issues in the Therapy of Patients with Cirrhosis due to NAFLD and NASH. Dig Dis 2015; 33:598-607. [PMID: 26159280 DOI: 10.1159/000375353] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the hepatic manifestation of the metabolic syndrome and comprises a liver disease spectrum ranging from steatosis to nonalcoholic steatohepatitis (NASH) with risk of progression to liver cirrhosis and hepatocellular carcinoma (HCC). Associated metabolic conditions and comorbidities such as obesity, diabetes and cardiovascular diseases are common and require concerted management. Adiponutrin (PNPLA3) variants may help to identify NAFLD patients at higher risk for liver disease progression towards advanced fibrosis and HCC. The therapeutic options in NAFLD/NASH include lifestyle modification, pharmacological treatment, bariatric surgery for patients with morbid obesity and treatment of complications of liver cirrhosis and HCC, including liver transplantation. Insulin sensitizers and antioxidative treatment strategies with vitamin E are among the best-established pharmacological approaches, but both drugs have long-term safety issues and there is limited evidence in cirrhotic patients. Treatment of concomitant/underlying metabolic conditions with statins or metformin may also have beneficial effects on portal hypertension, complications of liver cirrhosis and HCC prevention. The bile acid receptor FXR may be a promising novel therapeutic target for the treatment of NAFLD/NASH, fibrosis and portal hypertension, but the prognostic implications of associated changes in low- and high-density lipoprotein cholesterol require further studies. Morbidly obese NASH patients can benefit from bariatric surgery which may reduce liver fibrosis but carries a risk of decompensation in patients with advanced liver cirrhosis. When carefully selected, patients with NASH cirrhosis undergoing liver transplantation have a good outcome. This review summarizes recent progress in the management of patients with liver cirrhosis due to NASH.
Collapse
Affiliation(s)
- Stefan Traussnigg
- Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
1040
|
Farnesoid X receptor inhibits glucagon-like peptide-1 production by enteroendocrine L cells. Nat Commun 2015; 6:7629. [PMID: 26134028 PMCID: PMC4579574 DOI: 10.1038/ncomms8629] [Citation(s) in RCA: 289] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 05/25/2015] [Indexed: 02/02/2023] Open
Abstract
Bile acids are signalling molecules, which activate the transmembrane receptor TGR5 and the nuclear receptor FXR. BA sequestrants (BAS) complex bile acids in the intestinal lumen and decrease intestinal FXR activity. The BAS-BA complex also induces glucagon-like peptide-1 (GLP-1) production by L cells which potentiates β-cell glucose-induced insulin secretion. Whether FXR is expressed in L cells and controls GLP-1 production is unknown. Here, we show that FXR activation in L cells decreases proglucagon expression by interfering with the glucose-responsive factor Carbohydrate-Responsive Element Binding Protein (ChREBP) and GLP-1 secretion by inhibiting glycolysis. In vivo, FXR deficiency increases GLP-1 gene expression and secretion in response to glucose hence improving glucose metabolism. Moreover, treatment of ob/ob mice with the BAS colesevelam increases intestinal proglucagon gene expression and improves glycaemia in a FXR-dependent manner. These findings identify the FXR/GLP-1 pathway as a new mechanism of BA control of glucose metabolism and a pharmacological target for type 2 diabetes.
Collapse
|
1041
|
Poša M, Bjedov S, Škorić D, Sakač M. Micellization parameters (number average, aggregation number and critical micellar concentration) of bile salt 3 and 7 ethylidene derivatives: Role of the steroidal skeleton II. Biochim Biophys Acta Gen Subj 2015; 1850:1345-53. [DOI: 10.1016/j.bbagen.2015.03.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Revised: 03/23/2015] [Accepted: 03/24/2015] [Indexed: 01/22/2023]
|
1042
|
Abstract
OBJECTIVE Because the prevalence of obesity in children is increasing, the frequency of pediatric nonalcoholic fatty liver disease (NAFLD) is growing. A reliable noninvasive biomarker for monitoring progression of liver fibrosis would be useful. In cirrhotic persons serum bile acid (BA) levels are significantly elevated. We hypothesized that BA levels and composition in pediatric NAFLD vary depending on the stage of fibrosis. METHODS Children with NAFLD were compared with controls and classified by stages of fibrosis (NAFLD-F0, n = 27; NAFLD-F≥1, n = 65) based on liver-biopsy findings. Fasted metabolic and cholestasis status was assessed by several blood tests. BA profiles were measured by tandem mass spectrometry and compared with healthy controls (n = 105). RESULTS Compared with controls, all of the NAFLD patients were overweight and showed significantly elevated glucose, insulin, aspartate transaminase, and alanine transaminase levels. Total serum BAs were lower in nonfibrotic NAFLD children than in a control cohort (1.73 vs 3.6 μmol/L) because low glycine-conjugated BA levels were incompletely compensated by increases in taurine-conjugated or unconjugated BA. In patients with fibrotic NAFLD, BA levels were lower than in controls (2.45 vs 3.6 μmol/L) but higher than in nonfibrotic patients (2.45 vs 1.73 μmol/L), and the BA pattern resembled that of healthy controls. Fibroblast growth factor 19 levels were significantly lower in both NAFLD groups than in controls (P ≤ 0.001) and were positively correlated with ursodeoxycholic acid levels. CONCLUSIONS Our data indicate that serum BA levels decrease in early NAFLD and increase during progression to fibrosis. Given that BA levels are increased in cirrhotic adults, we postulate a continuous rise as NAFLD advances. BA may have a value as a noninvasive biomarker in pediatric NAFLD progression.
Collapse
|
1043
|
Steroidal scaffolds as FXR and GPBAR1 ligands: from chemistry to therapeutical application. Future Med Chem 2015; 7:1109-35. [DOI: 10.4155/fmc.15.54] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Bile acids (BAs) are experiencing a new life. Next to their ancestral roles in lipid digestion and solubilization, BAs are today recognized signaling molecules involved in many physiological functions. These signaling pathways involve the activation of metabolic nuclear receptors, mainly the BA sensor FXR, and the dedicated membrane G protein-coupled receptor, GPBAR1 (TGR5). As a consequence, the discovery of GPBAR1/FXR selective or dual modulators represents an important answer to the urgent demand of new pharmacological opportunity for several human diseases including dyslipidemia, cholestasis, nonalcoholic steatohepatitis, Type 2 diabetes and inflammation. Targeted oriented discovery of natural compounds and medicinal chemistry manipulation have allowed the development of promising drug candidates.
Collapse
|
1044
|
Park J, Al-Hilal TA, Jeong JH, Choi JU, Byun Y. Design, Synthesis, and Therapeutic Evaluation of Poly(acrylic acid)-tetraDOCA Conjugate as a Bile Acid Transporter Inhibitor. Bioconjug Chem 2015; 26:1597-605. [PMID: 26086474 DOI: 10.1021/acs.bioconjchem.5b00230] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Regulation of cholesterol and bile acid homeostasis has been attracting attention as a pharmaceutical target for the treatment of diseases, such as hypercholesterolaemia and type 2 diabetes. In recent years, small bile acid analogues have been developed for the purpose of apical sodium-dependent bile acid transporter (ASBT) inhibition. Here, we designed a novel hydrophilic ASBT inhibitor using oligomeric bile acid with a high affinity with ASBT. Polyacrylic acid-tetraDOCA conjugates (PATD) have the ability to bind to ASBT in order to induce hypocholesterolemic effects. Both the viability and the functionality of PATD were evaluated in vitro, showing that PATDs were effective in inhibiting the increases of cholesterol in the blood and oil in the liver induced by high fat diet (HFD). The results indicated that the newly developed biomaterials with oligomeric bile acids and a hydrophilic polymer are potent therapeutic agents for hyperlipidemia.
Collapse
Affiliation(s)
| | - Taslim A Al-Hilal
- ‡Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 136-791, South Korea
| | - Jee-Heon Jeong
- §College of Pharmacy, Yeungnam University, Gyeongsan 712-749, South Korea
| | | | | |
Collapse
|
1045
|
Chen X, Xu H, Ding L, Lou G, Liu Y, Yao Y, Chen L, Huang W, Fu X. Identification of miR-26a as a target gene of bile acid receptor GPBAR-1/TGR5. PLoS One 2015; 10:e0131294. [PMID: 26107166 PMCID: PMC4481113 DOI: 10.1371/journal.pone.0131294] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Accepted: 06/01/2015] [Indexed: 02/05/2023] Open
Abstract
GPBAR1/TGR5 is a G protein-coupled receptor of bile acids. TGR5 is known to regulate the BA homeostasis and energy metabolism. Recent studies highlight an important role of TGR5 in alleviating obesity and improving glucose regulation, however, the mechanism of which is still unclear. Here we report that TGR5 is involved in mediating the anti-obesity and anti-hyperglycemia effect of a natural compound, oleanolic acid. By comparing the miRNA profiles between wild type and TGR5-/- livers after OA treatment, we identified miR-26a as a novel downstream target gene of TGR5 activation. The expression of miR-26a in the liver was induced in a TGR5-dependent manner after feeding the mice with a bile acid diet. TGR5 activation strongly increased the expression of miR-26a in macrophages, including the Kupffer cells in the liver. We further demonstrated that JNK pathway was required for miR-26a induction by TGR5 activation. Interestingly, we located the TGR5-responsive DNA element to a proximal region of miR-26's promoter, which was independent of the transcription of its host genes. These results unravel a new mechanism by which bile acid receptor TGR5 activates a miRNA gene expression.
Collapse
Affiliation(s)
- Xiaosong Chen
- Department of Plastic Surgery, The Union Hospital of Fujian Medical University, 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Haixia Xu
- Division of Endocrinology and Metabolism, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, 610041, China
| | - Lili Ding
- Department of Diabetes Complications and Metabolism, Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, 1500 Duarte Road, Duarte, CA, 91010, United States of America
| | - Guiyu Lou
- Department of Diabetes Complications and Metabolism, Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, 1500 Duarte Road, Duarte, CA, 91010, United States of America
- Central Laboratory, Henan People's Hospital and Zhengzhou University People's Hospital, No.7 Wei Wu Road, Zhengzhou, Henan, 450003, China
| | - Yan Liu
- Department of Plastic Surgery, The Union Hospital of Fujian Medical University, 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Yalan Yao
- Division of Endocrinology and Metabolism, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, 610041, China
| | - Liangwan Chen
- Department of Plastic Surgery, The Union Hospital of Fujian Medical University, 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Wendong Huang
- Department of Diabetes Complications and Metabolism, Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, 1500 Duarte Road, Duarte, CA, 91010, United States of America
| | - Xianghui Fu
- Division of Endocrinology and Metabolism, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, 610041, China
| |
Collapse
|
1046
|
The gut microbiota of nonalcoholic fatty liver disease: current methods and their interpretation. Hepatol Int 2015; 9:406-15. [PMID: 26067771 PMCID: PMC4473019 DOI: 10.1007/s12072-015-9640-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 05/19/2015] [Indexed: 02/08/2023]
Abstract
The role of intestinal bacteria in the pathogenesis of nonalcoholic fatty liver disease is increasingly acknowledged. Recently developed microbial profiling techniques are beginning to shed light on the nature of gut microbiota alterations in nonalcoholic fatty liver disease. In this review, we summarize the gut microbiota composition changes that have been reported during different stages of human nonalcoholic fatty liver disease, and highlight the relation between bile acids and gut bacteria in this context. In addition, we discuss the different methodologies used in microbiota analyses as well as the interpretation of microbiota data. Whereas the currently available studies have provided useful information, future large-scale prospective studies with carefully phenotyped subjects and sequential sampling will be required to demonstrate a causal role of gut microbiota changes in the etiology of nonalcoholic fatty liver disease.
Collapse
|
1047
|
Kowal JM, Haanes KA, Christensen NM, Novak I. Bile acid effects are mediated by ATP release and purinergic signalling in exocrine pancreatic cells. Cell Commun Signal 2015; 13:28. [PMID: 26050734 PMCID: PMC4459444 DOI: 10.1186/s12964-015-0107-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 05/26/2015] [Indexed: 02/06/2023] Open
Abstract
Background In many cells, bile acids (BAs) have a multitude of effects, some of which may be mediated by specific receptors such the TGR5 or FXR receptors. In pancreas systemic BAs, as well as intra-ductal BAs from bile reflux, can affect pancreatic secretion. Extracellular ATP and purinergic signalling are other important regulators of similar secretory mechanisms in pancreas. The aim of our study was to elucidate whether there is interplay between ATP and BA signalling. Results Here we show that CDCA (chenodeoxycholic acid) caused fast and concentration-dependent ATP release from acini (AR42J) and duct cells (Capan-1). Taurine and glycine conjugated forms of CDCA had smaller effects on ATP release in Capan-1 cells. In duct monolayers, CDCA stimulated ATP release mainly from the luminal membrane; the releasing mechanisms involved both vesicular and non-vesicular secretion pathways. Duct cells were not depleted of intracellular ATP with CDCA, but acinar cells lost some ATP, as detected by several methods including ATP sensor AT1.03YEMK. In duct cells, CDCA caused reversible increase in the intracellular Ca2+ concentration [Ca2 +]i, which could be significantly inhibited by antagonists of purinergic receptors. The TGR5 receptor, expressed on the luminal side of pancreatic ducts, was not involved in ATP release and Ca2+ signals, but could stimulate Na+/Ca2+ exchange in some conditions. Conclusions CDCA evokes significant ATP release that can stimulate purinergic receptors, which in turn increase [Ca2+]i. The TGR5 receptor is not involved in these processes but can play a protective role at high intracellular Ca2+ conditions. We propose that purinergic signalling could be taken into consideration in other cells/organs, and thereby potentially explain some of the multifaceted effects of BAs. Electronic supplementary material The online version of this article (doi:10.1186/s12964-015-0107-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Justyna M Kowal
- Department of Biology, Section for Cell Biology and Physiology, August Krogh Building, University of Copenhagen, Universitetsparken 13, DK-2100, Copenhagen, Denmark.
| | - Kristian A Haanes
- Department of Biology, Section for Cell Biology and Physiology, August Krogh Building, University of Copenhagen, Universitetsparken 13, DK-2100, Copenhagen, Denmark. .,Present address: Department of Clinical Experimental Research, Glostrup Research Institute, Copenhagen University Hospital, Glostrup, Denmark.
| | - Nynne M Christensen
- Department of Biology, Section for Cell Biology and Physiology, August Krogh Building, University of Copenhagen, Universitetsparken 13, DK-2100, Copenhagen, Denmark.
| | - Ivana Novak
- Department of Biology, Section for Cell Biology and Physiology, August Krogh Building, University of Copenhagen, Universitetsparken 13, DK-2100, Copenhagen, Denmark.
| |
Collapse
|
1048
|
Abstract
TGR5 (Takeda G-protein-coupled receptor 5) [also known as GPBAR1 (G-protein-coupled bile acid receptor 1), M-BAR (membrane-type receptor for bile acids) or GPR131 (G-protein-coupled receptor 131)] is a G-protein-coupled receptor that was discovered as a bile acid receptor. TGR5 has specific roles in several tissues, among which are the regulation of energy expenditure, GLP-1 (glucagon-like peptide 1) secretion and gall bladder filling. An accumulating body of evidence now demonstrates that TGR5 also acts in a number of processes important in inflammation. Most striking in this context are several observations that TGR5 signalling curbs the inflammatory response of macrophages via interfering with NF-κB (nuclear factor κB) activity. In line with this, recent animal studies also suggest that TGR5 could be exploited as a potential target for intervention in a number of inflammation-driven diseases, including atherosclerosis. In the present paper, I review our current understanding of TGR5 with a strong focus on its potential as target for intervention in inflammation-driven diseases.
Collapse
|
1049
|
Prinz P, Hofmann T, Ahnis A, Elbelt U, Goebel-Stengel M, Klapp BF, Rose M, Stengel A. Plasma bile acids show a positive correlation with body mass index and are negatively associated with cognitive restraint of eating in obese patients. Front Neurosci 2015; 9:199. [PMID: 26089773 PMCID: PMC4452824 DOI: 10.3389/fnins.2015.00199] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 05/19/2015] [Indexed: 12/28/2022] Open
Abstract
Bile acids may be involved in the regulation of food intake and energy metabolism. The aim of the study was to investigate the association of plasma bile acids with body mass index (BMI) and the possible involvement of circulating bile acids in the modulation of physical activity and eating behavior. Blood was obtained in a group of hospitalized patients with normal weight (BMI 18.5–25 kg/m2), underweight (anorexia nervosa, BMI < 17.5 kg/m2) and overweight (obesity with BMI 30–40, 40–50 and >50 kg/m2, n = 14–15/group) and plasma bile acid concentrations assessed. Physical activity and plasma bile acids were measured in a group of patients with anorexia nervosa (BMI 14.6 ± 0.3 kg/m2, n = 43). Lastly, in a population of obese patients (BMI 48.5 ± 0.9 kg/m2, n = 85), psychometric parameters related to disordered eating and plasma bile acids were assessed. Plasma bile acids showed a positive correlation with BMI (r = 0.26, p = 0.03) in the population of patients with broad range of BMI (9–85 kg/m2, n = 74). No associations were observed between plasma bile acids and different parameters of physical activity in anorexic patients (p > 0.05). Plasma bile acids were negatively correlated with cognitive restraint of eating (r = −0.30, p = 0.008), while no associations were observed with other psychometric eating behavior-related parameters (p > 0.05) in obese patients. In conclusion, these data may point toward a role of bile acids in the regulation of body weight. Since plasma bile acids are negatively correlated with the cognitive restraint of eating in obese patients, this may represent a compensatory adaptation to prevent further overeating.
Collapse
Affiliation(s)
- Philip Prinz
- Division of General Internal and Psychosomatic Medicine, Charité Center for Internal Medicine and Dermatology, Charité-Universitätsmedizin Berlin Berlin, Germany
| | - Tobias Hofmann
- Division of General Internal and Psychosomatic Medicine, Charité Center for Internal Medicine and Dermatology, Charité-Universitätsmedizin Berlin Berlin, Germany
| | - Anne Ahnis
- Division of General Internal and Psychosomatic Medicine, Charité Center for Internal Medicine and Dermatology, Charité-Universitätsmedizin Berlin Berlin, Germany
| | - Ulf Elbelt
- Division of General Internal and Psychosomatic Medicine, Charité Center for Internal Medicine and Dermatology, Charité-Universitätsmedizin Berlin Berlin, Germany ; Division for Endocrinology, Diabetes, and Nutrition, Charité Center for Internal Medicine with Gastroenterology and Nephrology, Charité-Universitätsmedizin Berlin Berlin, Germany
| | - Miriam Goebel-Stengel
- Department of Internal Medicine and Institute of Neurogastroenterology, Martin-Luther-Krankenhaus Berlin, Germany
| | - Burghard F Klapp
- Division of General Internal and Psychosomatic Medicine, Charité Center for Internal Medicine and Dermatology, Charité-Universitätsmedizin Berlin Berlin, Germany
| | - Matthias Rose
- Division of General Internal and Psychosomatic Medicine, Charité Center for Internal Medicine and Dermatology, Charité-Universitätsmedizin Berlin Berlin, Germany
| | - Andreas Stengel
- Division of General Internal and Psychosomatic Medicine, Charité Center for Internal Medicine and Dermatology, Charité-Universitätsmedizin Berlin Berlin, Germany
| |
Collapse
|
1050
|
Abstract
Bariatric surgery is arguably the most effective therapy for weight loss, and Rouen-Y gastric bypass (RYGB) is considered the "gold-standard" procedure. However, sleeve gastrectomy (SG) surgery has become more prevalent in recent years and it is unclear if weight loss differences occur between these procedures. Herein, we discuss evidence from randomized clinical trials comparing the effectiveness of RYGB and SG on weight loss. Moreover, we highlight gut hormones (e.g., GLP-1, ghrelin, bile acids, etc.) as potentially important mechanisms that contribute to the durability of decreased appetite and opposed fat storage following RYGB and SG. Collectively, although a subtle (∼ 3-5 kg) weight loss difference may exist in favor of RYGB up to 3 years post-operation, it appears that RYGB and SG induce comparable weight loss and changes in gut physiology that parallel reduced disease risk. These findings are clinically relevant for optimizing treatment strategies that combat obesity-related diabetes and cardiovascular disease.
Collapse
Affiliation(s)
- Steven K Malin
- Department of Kinesiology, University of Virginia, 210 Memorial Gymnasium, Charlottesville, VA, USA.
- Division of Endocrinology and Metabolism, University of Virginia, 210 Memorial Gymnasium, Charlottesville, VA, USA.
| | - Sangeeta R Kashyap
- Department of Endocrinology, Diabetes, and Metabolism, Cleveland Clinic, 9500 Euclid Ave (NE40), Cleveland, OH, 44195, USA.
| |
Collapse
|