1051
|
Nuevos agentes terapéuticos para la diabetes tipo 2. Med Clin (Barc) 2015; 144:560-5. [DOI: 10.1016/j.medcli.2014.03.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 03/13/2014] [Indexed: 01/10/2023]
|
1052
|
Abstract
Obesity is a multifactorial disorder that results in excessive accumulation of adipose tissue. Although obesity is caused by alterations in the energy consumption/expenditure balance, the factors promoting this disequilibrium are incompletely understood. The rapid development of new technologies and analysis strategies to decode the gut microbiota composition and metabolic pathways has opened a door into the complexity of the guest-host interactions between the gut microbiota and its human host in health and in disease. Pivotal studies have demonstrated that manipulation of the gut microbiota and its metabolic pathways can affect host's adiposity and metabolism. These observations have paved the way for further assessment of the mechanisms underlying these changes. In this review we summarize the current evidence for possible mechanisms underlying gut microbiota induced obesity. The review addresses some well-known effects of the gut microbiota on energy harvesting and changes in metabolic machinery, on metabolic and immune interactions and on possible changes in brain function and behavior. Although there is limited understanding on the symbiotic relationship between us and our gut microbiome, and how disturbances of this relationship affects our health, there is compelling evidence for an important role of the gut microbiota in the development and perpetuation of obesity.
Collapse
Affiliation(s)
- Claudia Sanmiguel
- Oppenheimer Center for Neurobiology of Stress, Los Angeles, CA
- Department of Medicine, Los Angeles, CA
| | - Arpana Gupta
- Oppenheimer Center for Neurobiology of Stress, Los Angeles, CA
- Department of Medicine, Los Angeles, CA
| | - Emeran A. Mayer
- Oppenheimer Center for Neurobiology of Stress, Los Angeles, CA
- Department of Medicine, Los Angeles, CA
- Department of Physiology, Los Angeles, CA
- Department of Psychiatry, Los Angeles, CA
- UCLA CURE Digestive Diseases Research Center, Los Angeles, CA
| |
Collapse
|
1053
|
Abstract
Systemic inflammation resulting from dysfunction of white adipose tissue (WAT) accelerates the pathologies of diabetes and cardiovascular diseases. In contrast to WAT, brown adipose tissue (BAT) is abundant in mitochondria that produce heat by uncoupling respiratory chain process of ATP synthesis. Besides BAT's role in thermogenesis, accumulating evidence has shown that it is involved in regulating systemic metabolism. Studies have analyzed the "browning" processes of WAT as a means to combat obesity, whereas few studies have focused on the impact and molecular mechanisms that contribute to obesity-linked BAT dysfunction--a process that is associated with the "whitening" of this tissue. Compared to WAT, a dense vascular network is required to support the high energy consumption of BAT. Recently, vascular rarefaction was shown to be a significant causal factor in the whitening of BAT in mouse models. Vascular insufficiency leads to mitochondrial dysfunction and loss in BAT and contributes to systemic insulin resistance. These data suggest that BAT "whitening," resulting from vascular dysfunction, can impact obesity and obesity-linked diseases. Conversely, agents that promote BAT function could have utility in the treatment of these conditions.
Collapse
Affiliation(s)
- Ippei Shimizu
- Department of Cardiovascular Biology and Medicine, Division of Molecular Aging and Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8510, Japan.
| | - Kenneth Walsh
- Molecular Cardiology and Whitaker Cardiovascular Institute, Boston University School of Medicine, 715 Albany Street, W611, Boston, MA, 02118, USA
| |
Collapse
|
1054
|
Jourdainne V, Péan N, Doignon I, Humbert L, Rainteau D, Tordjmann T. The Bile Acid Receptor TGR5 and Liver Regeneration. Dig Dis 2015; 33:319-26. [PMID: 26045264 DOI: 10.1159/000371668] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Most of the literature on the bile acid (BA) membrane receptor TGR5 is dedicated to its potential role in the metabolic syndrome, through its regulatory impact on energy expenditure, insulin and GLP-1 secretion, and inflammatory processes. While the receptor was cloned in 2002, very little data are available on TGR5 functions in the normal and diseased liver. However, TGR5 is highly expressed in Kupffer cells and liver endothelial cells, and is particularly enriched in the biliary tract [cholangiocytes and gallbladder (GB) smooth muscle cells]. We recently demonstrated that TGR5 has a crucial protective impact on the liver in case of BA overload, including after partial hepatectomy. KEY MESSAGES TGR5-KO mice after PH exhibited periportal bile infarcts, excessive hepatic inflammation and defective adaptation of biliary composition (bicarbonate and chloride). Most importantly, TGR5-KO mice had a more hydrophobic BA pool, with more secondary BA than WT animals, suggesting that TGR5-KO bile may be harmful for the liver, mainly in situations of BA overload. As GB is both the tissue displaying the highest level of TGR5 expression and a crucial physiological site for the regulation of BA pool hydrophobicity by reducing secondary BA, we investigated whether TGR5 may control BA pool composition through an impact on GB. Preliminary data suggest that in the absence of TGR5, reduced GB filling dampens the cholecystohepatic shunt, resulting in more secondary BA, more hydrophobic BA pool and extensive liver injury in case of BA overload. CONCLUSIONS In the setting of BA overload, TGR5 is protective of the liver through the regulation of not only secretory and inflammatory processes, but also through the control of BA pool composition, at least in part by targeting the GB. Thereby, TGR5 appears to be crucial for protecting the regenerating liver from BA overload.
Collapse
|
1055
|
Zhou H, Zhou S, Gao J, Zhang G, Lu Y, Owyang C. Upregulation of bile acid receptor TGR5 and nNOS in gastric myenteric plexus is responsible for delayed gastric emptying after chronic high-fat feeding in rats. Am J Physiol Gastrointest Liver Physiol 2015; 308:G863-73. [PMID: 25540233 PMCID: PMC4437020 DOI: 10.1152/ajpgi.00380.2014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 12/19/2014] [Indexed: 01/31/2023]
Abstract
Chronic high-fat feeding is associated with functional dyspepsia and delayed gastric emptying. We hypothesize that high-fat feeding upregulates gastric neuronal nitric oxide synthase (nNOS) expression, resulting in delayed gastric emptying. We propose this is mediated by increased bile acid action on bile acid receptor 1 (TGR5) located on nNOS gastric neurons. To test this hypothesis, rats were fed regular chow or a high-fat diet for 2 wk. Rats fed the high-fat diet were subjected to concurrent feeding with oral cholestyramine or terminal ileum resection. TGR5 and nNOS expression in gastric tissue was measured by immunohistochemistry, PCR, and Western blot. Gastric motility was assessed by organ bath and solid-phase gastric emptying studies. The 2-wk high-fat diet caused a significant increase in neurons coexpressing nNOS and TGR5 in the gastric myenteric plexus and an increase in nNOS and TGR5 gene expression, 67 and 111%, respectively. Enhanced nonadrenergic, noncholinergic (NANC) relaxation, deoxycholic acid (DCA)-induced inhibition in fundic tissue, and a 26% delay in gastric emptying accompanied these changes. A 24-h incubation of whole-mount gastric fundus with DCA resulted in increased nNOS and TGR5 protein expression, 41 and 37%, respectively. Oral cholestyramine and terminal ileum resection restored the enhanced gastric relaxation, as well as the elevated nNOS and TGR5 expression evoked by high-fat feeding. Cholestyramine also prevented the delay in gastric emptying. We conclude that increased levels of circulatory bile acids induced by high-fat feeding upregulate nNOS and TGR5 expression in the gastric myenteric plexus, resulting in enhanced NANC relaxation and delayed gastric emptying.
Collapse
Affiliation(s)
- Hui Zhou
- 1Division of Gastroenterology, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan; and ,2Department of Gastroenterology, Shanghai First People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Shiyi Zhou
- 1Division of Gastroenterology, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan; and
| | - Jun Gao
- 1Division of Gastroenterology, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan; and
| | - Guanpo Zhang
- 1Division of Gastroenterology, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan; and
| | - Yuanxu Lu
- 1Division of Gastroenterology, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan; and
| | - Chung Owyang
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan; and
| |
Collapse
|
1056
|
Song Y, Xu C, Shao S, Liu J, Xing W, Xu J, Qin C, Li C, Hu B, Yi S, Xia X, Zhang H, Zhang X, Wang T, Pan W, Yu C, Wang Q, Lin X, Wang L, Gao L, Zhao J. Thyroid-stimulating hormone regulates hepatic bile acid homeostasis via SREBP-2/HNF-4α/CYP7A1 axis. J Hepatol 2015; 62:1171-9. [PMID: 25533663 DOI: 10.1016/j.jhep.2014.12.006] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 12/03/2014] [Accepted: 12/03/2014] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS Bile acids (BAs) play a crucial role in dietary fat digestion and in the regulation of lipid, glucose, and energy metabolism. Thyroid-stimulating hormone (TSH) is a hormone produced by the anterior pituitary gland that directly regulates several metabolic pathways. However, the impact of TSH on BA homeostasis remains largely unknown. METHODS We analyzed serum BA and TSH levels in healthy volunteers under strict control of caloric intake. Thyroidectomized rats were administered thyroxine and injected with different doses of TSH. Tshr(-/-) mice were supplemented with thyroxine, and C57BL/6 mice were injected with Tshr-siRNA via the tail vein. The serum BA levels, BA pool size, and fecal BA excretion rate were measured. The regulation of SREBP-2, HNF-4α, and CYP7A1 by TSH were analyzed using luciferase reporter, RNAi, EMSA, and CHIP assays. RESULTS A negative correlation was observed between the serum levels of TSH and the serum BA levels in healthy volunteers. TSH administration led to a decrease in BA content and CYP7A1 activity in thyroidectomized rats supplemented with thyroxine. When Tshr was silenced in mice, the BA pool size, fecal BA excretion rate, and serum BA levels all increased. Additionally, we found that HNF-4α acts as a critical molecule through which TSH represses CYP7A1 activity. We further confirmed that the accumulation of mature SREBP-2 protein could impair the capacity of nuclear HNF-4α to bind to the CYP7A1 promoter, a mechanism that appears to mediate the effects of TSH. CONCLUSIONS TSH represses hepatic BA synthesis via a SREBP-2/HNF-4α/CYP7A1 signaling pathway. This finding strongly supports the notion that TSH is an important pathophysiological regulator of liver BA homeostasis independently of thyroid hormones.
Collapse
Affiliation(s)
- Yongfeng Song
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
| | - Chao Xu
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
| | - Shanshan Shao
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
| | - Jun Liu
- Department of Organ Transplantation Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Wanjia Xing
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
| | - Jin Xu
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
| | - Chengkun Qin
- Department of General Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Chunyou Li
- Department of Organ Transplantation Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Baoxiang Hu
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
| | - Shounan Yi
- Center for Transplant and Renal Research, Westmead Millennium Institute, University of Sydney at Westmead Hospital, Sydney, Australia
| | - Xuefeng Xia
- Genomic Medicine and Center for Diabetes Research, The Methodist Hospital Research Institute, Weill Cornell Medical College, Houston, TX 77030, USA
| | - Haiqing Zhang
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
| | - Xiujuan Zhang
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
| | - Tingting Wang
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
| | - Wenfei Pan
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
| | - Chunxiao Yu
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
| | - Qiangxiu Wang
- Department of Pathology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Xiaoyan Lin
- Department of Pathology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Laicheng Wang
- Scientific Center, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Ling Gao
- Scientific Center, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China.
| | - Jiajun Zhao
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China; Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China.
| |
Collapse
|
1057
|
Dutia R, Embrey M, O’Brien S, Haeusler RA, Agénor KK, Homel P, McGinty J, Vincent RP, Alaghband-Zadeh J, Staels B, le Roux CW, Yu J, Laferrère B. Temporal changes in bile acid levels and 12α-hydroxylation after Roux-en-Y gastric bypass surgery in type 2 diabetes. Int J Obes (Lond) 2015; 39:806-13. [PMID: 25599611 PMCID: PMC4422767 DOI: 10.1038/ijo.2015.1] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Revised: 11/26/2014] [Accepted: 12/25/2014] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Gastric bypass surgery (GBP) leads to sustained weight loss and significant improvement in type 2 diabetes (T2DM). Bile acids (BAs), signaling molecules which influence glucose metabolism, are a potential mediator for the improvement in T2DM after GBP. This study sought to investigate the effect of GBP on BA levels and composition in individuals with T2DM. METHODS Plasma BA levels and composition and fibroblast growth factor (FGF)-19 levels were measured during fasting and in response to an oral glucose load before and at 1 month and 2 years post GBP in 13 severely obese women with T2DM. RESULTS A striking temporal change in BA levels and composition was observed after GBP. During the fasted state, BA concentrations were generally reduced at 1 month, but increased 2 years post GBP. Postprandial BA levels were unchanged 1 month post GBP, but an exaggerated postprandial peak was observed 2 years after the surgery. A significant increase in the 12α-hydroxylated/non12α-hydroxylated BA ratio during fasting and postprandially at 2 years, but not 1 month, post GBP was observed. Significant correlations between BAs vs FGF-19, body weight, the incretin effect and peptide YY (PYY) were also found. CONCLUSIONS This study provides evidence that GBP temporally modifies the concentration and composition of circulating BAs in individuals with T2DM, and suggests that BAs may be linked to the improvement in T2DM after GBP.
Collapse
Affiliation(s)
- R Dutia
- New York Obesity Nutrition Research Center, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - M Embrey
- New York Obesity Nutrition Research Center, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - S O’Brien
- New York Obesity Nutrition Research Center, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - RA Haeusler
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - KK Agénor
- New York Obesity Nutrition Research Center, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - P Homel
- Department of Medicine, Albert Einstein School of Medicine, New York, NY, USA
| | - J McGinty
- Division of Bariatric and Minimally Invasive Surgery, Department of Surgery, Mount Sinai, St. Luke’s Roosevelt Hospitals, New York, NY, USA
| | - RP Vincent
- Department of Clinical Biochemistry, King's College Hospital NHS Foundation Trust Denmark Hill, London, UK
| | - J Alaghband-Zadeh
- Department of Clinical Biochemistry, Guy's & St Thomas' NHS Foundation Trust, London, UK
| | - B Staels
- European Genomic Institute for Diabetes (EGID), Université Lille 2, Inserm UMR 1011, Institut Pasteur de Lille, Lille, France
| | - CW le Roux
- Diabetes Complications Research Center, Conway Institute, University College, Dublin, Ireland
| | - J Yu
- New York Obesity Nutrition Research Center, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - B Laferrère
- New York Obesity Nutrition Research Center, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
| |
Collapse
|
1058
|
Role of Intestinal Microbiome in Lipid and Glucose Metabolism in Diabetes Mellitus. Clin Ther 2015; 37:1172-7. [PMID: 25922340 DOI: 10.1016/j.clinthera.2015.03.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 02/19/2015] [Accepted: 03/02/2015] [Indexed: 12/15/2022]
Abstract
PURPOSE The contribution of intestinal bacterial strains (gut microbiota) in human metabolism and obesity is being increasingly recognized. The goal of this article was to provide a commentary on the clinical usefulness of these data. METHODS We performed a review of the currently available articles on PubMed. FINDINGS Because most of the data are based on germ-free animal research, translation to human disease may be difficult. However, changes in the intestinal bacterial composition and subsequent altered diversity have been associated with the presence of chronic low-grade inflammation, a known feature of insulin resistance and type 2 diabetes mellitus. IMPLICATIONS It is still not proven whether intestinal bacteria play a causal role in glucose and lipid metabolism. Intervention studies including fecal transplantation and supplementation of single bacterial strains in humans might provide more insight. Moreover, prospective cohorts of healthy subjects using fecal samples collected at baseline can help to identify causally involved specific intestinal bacterial strains that drive aberrant human metabolism. Ultimately, it would be a great asset if potential diagnostic and therapeutic targets could be derived from this novel player in human cardiometabolism.
Collapse
|
1059
|
Cheng S, Larson MG, McCabe EL, Murabito JM, Rhee EP, Ho JE, Jacques PF, Ghorbani A, Magnusson M, Souza AL, Deik AA, Pierce KA, Bullock K, O'Donnell CJ, Melander O, Clish CB, Vasan RS, Gerszten RE, Wang TJ. Distinct metabolomic signatures are associated with longevity in humans. Nat Commun 2015; 6:6791. [PMID: 25864806 PMCID: PMC4396657 DOI: 10.1038/ncomms7791] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 02/27/2015] [Indexed: 01/07/2023] Open
Abstract
Alterations in metabolism influence lifespan in experimental models, but data in humans are lacking. Here we use liquid chromatography/mass spectrometry to quantify 217 plasma metabolites and examine their relation to longevity in a large cohort of men and women followed for up to 20 years. We find that, higher concentrations of the citric acid cycle intermediate, isocitrate, and the bile acid, taurocholate, are associated with lower odds of longevity, defined as attaining 80 years of age. Higher concentrations of isocitrate, but not taurocholate, are also associated with worse cardiovascular health at baseline, as well as risk of future cardiovascular disease and death. None of the metabolites identified are associated with cancer risk. Our findings suggest that some, but not all, metabolic pathways related to human longevity are linked to the risk of common causes of death.
Collapse
Affiliation(s)
- Susan Cheng
- Framingham Heart Study of the National Heart, Lung and Blood Institute and Boston University School of Medicine, Framingham, MA (SC, MGL, JMM, JEH, CJO, RSV, TJW); Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (SC); Department of Mathematics and Statistics, Boston University, Boston, MA (MGL); Department of Biostatistics, Boston University School of Public Health, Boston, MA (ELM); Cardiology Division (JEH, AG, CJO, REG), Cardiovascular Research Center (REG), and Renal Division (EPR), Massachusetts General Hospital, Harvard Medical School, Boston, MA; General Internal Medicine (JMM), Cardiology (JEH, RSV), and Preventive Medicine (RSV), Department of Medicine, Boston University School of Medicine, Boston, MA; Jean Mayer US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA (PFJ); Department of Clinical Sciences, Lund University, Malmö (MM, OM); Broad Institute of MIT and Harvard, Cambridge, MA (ALS, AAD, KAP, KB, CBC, REG); National Heart, Lung & Blood Institute Division of Intramural Research, Bethesda, MD (CJO); and, Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (TJW)
| | - Martin G Larson
- Framingham Heart Study of the National Heart, Lung and Blood Institute and Boston University School of Medicine, Framingham, MA (SC, MGL, JMM, JEH, CJO, RSV, TJW); Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (SC); Department of Mathematics and Statistics, Boston University, Boston, MA (MGL); Department of Biostatistics, Boston University School of Public Health, Boston, MA (ELM); Cardiology Division (JEH, AG, CJO, REG), Cardiovascular Research Center (REG), and Renal Division (EPR), Massachusetts General Hospital, Harvard Medical School, Boston, MA; General Internal Medicine (JMM), Cardiology (JEH, RSV), and Preventive Medicine (RSV), Department of Medicine, Boston University School of Medicine, Boston, MA; Jean Mayer US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA (PFJ); Department of Clinical Sciences, Lund University, Malmö (MM, OM); Broad Institute of MIT and Harvard, Cambridge, MA (ALS, AAD, KAP, KB, CBC, REG); National Heart, Lung & Blood Institute Division of Intramural Research, Bethesda, MD (CJO); and, Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (TJW)
| | - Elizabeth L McCabe
- Framingham Heart Study of the National Heart, Lung and Blood Institute and Boston University School of Medicine, Framingham, MA (SC, MGL, JMM, JEH, CJO, RSV, TJW); Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (SC); Department of Mathematics and Statistics, Boston University, Boston, MA (MGL); Department of Biostatistics, Boston University School of Public Health, Boston, MA (ELM); Cardiology Division (JEH, AG, CJO, REG), Cardiovascular Research Center (REG), and Renal Division (EPR), Massachusetts General Hospital, Harvard Medical School, Boston, MA; General Internal Medicine (JMM), Cardiology (JEH, RSV), and Preventive Medicine (RSV), Department of Medicine, Boston University School of Medicine, Boston, MA; Jean Mayer US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA (PFJ); Department of Clinical Sciences, Lund University, Malmö (MM, OM); Broad Institute of MIT and Harvard, Cambridge, MA (ALS, AAD, KAP, KB, CBC, REG); National Heart, Lung & Blood Institute Division of Intramural Research, Bethesda, MD (CJO); and, Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (TJW)
| | - Joanne M Murabito
- Framingham Heart Study of the National Heart, Lung and Blood Institute and Boston University School of Medicine, Framingham, MA (SC, MGL, JMM, JEH, CJO, RSV, TJW); Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (SC); Department of Mathematics and Statistics, Boston University, Boston, MA (MGL); Department of Biostatistics, Boston University School of Public Health, Boston, MA (ELM); Cardiology Division (JEH, AG, CJO, REG), Cardiovascular Research Center (REG), and Renal Division (EPR), Massachusetts General Hospital, Harvard Medical School, Boston, MA; General Internal Medicine (JMM), Cardiology (JEH, RSV), and Preventive Medicine (RSV), Department of Medicine, Boston University School of Medicine, Boston, MA; Jean Mayer US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA (PFJ); Department of Clinical Sciences, Lund University, Malmö (MM, OM); Broad Institute of MIT and Harvard, Cambridge, MA (ALS, AAD, KAP, KB, CBC, REG); National Heart, Lung & Blood Institute Division of Intramural Research, Bethesda, MD (CJO); and, Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (TJW)
| | - Eugene P Rhee
- Framingham Heart Study of the National Heart, Lung and Blood Institute and Boston University School of Medicine, Framingham, MA (SC, MGL, JMM, JEH, CJO, RSV, TJW); Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (SC); Department of Mathematics and Statistics, Boston University, Boston, MA (MGL); Department of Biostatistics, Boston University School of Public Health, Boston, MA (ELM); Cardiology Division (JEH, AG, CJO, REG), Cardiovascular Research Center (REG), and Renal Division (EPR), Massachusetts General Hospital, Harvard Medical School, Boston, MA; General Internal Medicine (JMM), Cardiology (JEH, RSV), and Preventive Medicine (RSV), Department of Medicine, Boston University School of Medicine, Boston, MA; Jean Mayer US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA (PFJ); Department of Clinical Sciences, Lund University, Malmö (MM, OM); Broad Institute of MIT and Harvard, Cambridge, MA (ALS, AAD, KAP, KB, CBC, REG); National Heart, Lung & Blood Institute Division of Intramural Research, Bethesda, MD (CJO); and, Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (TJW)
| | - Jennifer E Ho
- Framingham Heart Study of the National Heart, Lung and Blood Institute and Boston University School of Medicine, Framingham, MA (SC, MGL, JMM, JEH, CJO, RSV, TJW); Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (SC); Department of Mathematics and Statistics, Boston University, Boston, MA (MGL); Department of Biostatistics, Boston University School of Public Health, Boston, MA (ELM); Cardiology Division (JEH, AG, CJO, REG), Cardiovascular Research Center (REG), and Renal Division (EPR), Massachusetts General Hospital, Harvard Medical School, Boston, MA; General Internal Medicine (JMM), Cardiology (JEH, RSV), and Preventive Medicine (RSV), Department of Medicine, Boston University School of Medicine, Boston, MA; Jean Mayer US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA (PFJ); Department of Clinical Sciences, Lund University, Malmö (MM, OM); Broad Institute of MIT and Harvard, Cambridge, MA (ALS, AAD, KAP, KB, CBC, REG); National Heart, Lung & Blood Institute Division of Intramural Research, Bethesda, MD (CJO); and, Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (TJW)
| | - Paul F Jacques
- Framingham Heart Study of the National Heart, Lung and Blood Institute and Boston University School of Medicine, Framingham, MA (SC, MGL, JMM, JEH, CJO, RSV, TJW); Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (SC); Department of Mathematics and Statistics, Boston University, Boston, MA (MGL); Department of Biostatistics, Boston University School of Public Health, Boston, MA (ELM); Cardiology Division (JEH, AG, CJO, REG), Cardiovascular Research Center (REG), and Renal Division (EPR), Massachusetts General Hospital, Harvard Medical School, Boston, MA; General Internal Medicine (JMM), Cardiology (JEH, RSV), and Preventive Medicine (RSV), Department of Medicine, Boston University School of Medicine, Boston, MA; Jean Mayer US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA (PFJ); Department of Clinical Sciences, Lund University, Malmö (MM, OM); Broad Institute of MIT and Harvard, Cambridge, MA (ALS, AAD, KAP, KB, CBC, REG); National Heart, Lung & Blood Institute Division of Intramural Research, Bethesda, MD (CJO); and, Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (TJW)
| | - Anahita Ghorbani
- Framingham Heart Study of the National Heart, Lung and Blood Institute and Boston University School of Medicine, Framingham, MA (SC, MGL, JMM, JEH, CJO, RSV, TJW); Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (SC); Department of Mathematics and Statistics, Boston University, Boston, MA (MGL); Department of Biostatistics, Boston University School of Public Health, Boston, MA (ELM); Cardiology Division (JEH, AG, CJO, REG), Cardiovascular Research Center (REG), and Renal Division (EPR), Massachusetts General Hospital, Harvard Medical School, Boston, MA; General Internal Medicine (JMM), Cardiology (JEH, RSV), and Preventive Medicine (RSV), Department of Medicine, Boston University School of Medicine, Boston, MA; Jean Mayer US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA (PFJ); Department of Clinical Sciences, Lund University, Malmö (MM, OM); Broad Institute of MIT and Harvard, Cambridge, MA (ALS, AAD, KAP, KB, CBC, REG); National Heart, Lung & Blood Institute Division of Intramural Research, Bethesda, MD (CJO); and, Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (TJW)
| | - Martin Magnusson
- Framingham Heart Study of the National Heart, Lung and Blood Institute and Boston University School of Medicine, Framingham, MA (SC, MGL, JMM, JEH, CJO, RSV, TJW); Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (SC); Department of Mathematics and Statistics, Boston University, Boston, MA (MGL); Department of Biostatistics, Boston University School of Public Health, Boston, MA (ELM); Cardiology Division (JEH, AG, CJO, REG), Cardiovascular Research Center (REG), and Renal Division (EPR), Massachusetts General Hospital, Harvard Medical School, Boston, MA; General Internal Medicine (JMM), Cardiology (JEH, RSV), and Preventive Medicine (RSV), Department of Medicine, Boston University School of Medicine, Boston, MA; Jean Mayer US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA (PFJ); Department of Clinical Sciences, Lund University, Malmö (MM, OM); Broad Institute of MIT and Harvard, Cambridge, MA (ALS, AAD, KAP, KB, CBC, REG); National Heart, Lung & Blood Institute Division of Intramural Research, Bethesda, MD (CJO); and, Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (TJW)
| | - Amanda L Souza
- Framingham Heart Study of the National Heart, Lung and Blood Institute and Boston University School of Medicine, Framingham, MA (SC, MGL, JMM, JEH, CJO, RSV, TJW); Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (SC); Department of Mathematics and Statistics, Boston University, Boston, MA (MGL); Department of Biostatistics, Boston University School of Public Health, Boston, MA (ELM); Cardiology Division (JEH, AG, CJO, REG), Cardiovascular Research Center (REG), and Renal Division (EPR), Massachusetts General Hospital, Harvard Medical School, Boston, MA; General Internal Medicine (JMM), Cardiology (JEH, RSV), and Preventive Medicine (RSV), Department of Medicine, Boston University School of Medicine, Boston, MA; Jean Mayer US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA (PFJ); Department of Clinical Sciences, Lund University, Malmö (MM, OM); Broad Institute of MIT and Harvard, Cambridge, MA (ALS, AAD, KAP, KB, CBC, REG); National Heart, Lung & Blood Institute Division of Intramural Research, Bethesda, MD (CJO); and, Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (TJW)
| | - Amy A Deik
- Framingham Heart Study of the National Heart, Lung and Blood Institute and Boston University School of Medicine, Framingham, MA (SC, MGL, JMM, JEH, CJO, RSV, TJW); Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (SC); Department of Mathematics and Statistics, Boston University, Boston, MA (MGL); Department of Biostatistics, Boston University School of Public Health, Boston, MA (ELM); Cardiology Division (JEH, AG, CJO, REG), Cardiovascular Research Center (REG), and Renal Division (EPR), Massachusetts General Hospital, Harvard Medical School, Boston, MA; General Internal Medicine (JMM), Cardiology (JEH, RSV), and Preventive Medicine (RSV), Department of Medicine, Boston University School of Medicine, Boston, MA; Jean Mayer US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA (PFJ); Department of Clinical Sciences, Lund University, Malmö (MM, OM); Broad Institute of MIT and Harvard, Cambridge, MA (ALS, AAD, KAP, KB, CBC, REG); National Heart, Lung & Blood Institute Division of Intramural Research, Bethesda, MD (CJO); and, Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (TJW)
| | - Kerry A Pierce
- Framingham Heart Study of the National Heart, Lung and Blood Institute and Boston University School of Medicine, Framingham, MA (SC, MGL, JMM, JEH, CJO, RSV, TJW); Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (SC); Department of Mathematics and Statistics, Boston University, Boston, MA (MGL); Department of Biostatistics, Boston University School of Public Health, Boston, MA (ELM); Cardiology Division (JEH, AG, CJO, REG), Cardiovascular Research Center (REG), and Renal Division (EPR), Massachusetts General Hospital, Harvard Medical School, Boston, MA; General Internal Medicine (JMM), Cardiology (JEH, RSV), and Preventive Medicine (RSV), Department of Medicine, Boston University School of Medicine, Boston, MA; Jean Mayer US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA (PFJ); Department of Clinical Sciences, Lund University, Malmö (MM, OM); Broad Institute of MIT and Harvard, Cambridge, MA (ALS, AAD, KAP, KB, CBC, REG); National Heart, Lung & Blood Institute Division of Intramural Research, Bethesda, MD (CJO); and, Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (TJW)
| | - Kevin Bullock
- Framingham Heart Study of the National Heart, Lung and Blood Institute and Boston University School of Medicine, Framingham, MA (SC, MGL, JMM, JEH, CJO, RSV, TJW); Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (SC); Department of Mathematics and Statistics, Boston University, Boston, MA (MGL); Department of Biostatistics, Boston University School of Public Health, Boston, MA (ELM); Cardiology Division (JEH, AG, CJO, REG), Cardiovascular Research Center (REG), and Renal Division (EPR), Massachusetts General Hospital, Harvard Medical School, Boston, MA; General Internal Medicine (JMM), Cardiology (JEH, RSV), and Preventive Medicine (RSV), Department of Medicine, Boston University School of Medicine, Boston, MA; Jean Mayer US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA (PFJ); Department of Clinical Sciences, Lund University, Malmö (MM, OM); Broad Institute of MIT and Harvard, Cambridge, MA (ALS, AAD, KAP, KB, CBC, REG); National Heart, Lung & Blood Institute Division of Intramural Research, Bethesda, MD (CJO); and, Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (TJW)
| | - Christopher J O'Donnell
- Framingham Heart Study of the National Heart, Lung and Blood Institute and Boston University School of Medicine, Framingham, MA (SC, MGL, JMM, JEH, CJO, RSV, TJW); Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (SC); Department of Mathematics and Statistics, Boston University, Boston, MA (MGL); Department of Biostatistics, Boston University School of Public Health, Boston, MA (ELM); Cardiology Division (JEH, AG, CJO, REG), Cardiovascular Research Center (REG), and Renal Division (EPR), Massachusetts General Hospital, Harvard Medical School, Boston, MA; General Internal Medicine (JMM), Cardiology (JEH, RSV), and Preventive Medicine (RSV), Department of Medicine, Boston University School of Medicine, Boston, MA; Jean Mayer US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA (PFJ); Department of Clinical Sciences, Lund University, Malmö (MM, OM); Broad Institute of MIT and Harvard, Cambridge, MA (ALS, AAD, KAP, KB, CBC, REG); National Heart, Lung & Blood Institute Division of Intramural Research, Bethesda, MD (CJO); and, Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (TJW)
| | - Olle Melander
- Framingham Heart Study of the National Heart, Lung and Blood Institute and Boston University School of Medicine, Framingham, MA (SC, MGL, JMM, JEH, CJO, RSV, TJW); Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (SC); Department of Mathematics and Statistics, Boston University, Boston, MA (MGL); Department of Biostatistics, Boston University School of Public Health, Boston, MA (ELM); Cardiology Division (JEH, AG, CJO, REG), Cardiovascular Research Center (REG), and Renal Division (EPR), Massachusetts General Hospital, Harvard Medical School, Boston, MA; General Internal Medicine (JMM), Cardiology (JEH, RSV), and Preventive Medicine (RSV), Department of Medicine, Boston University School of Medicine, Boston, MA; Jean Mayer US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA (PFJ); Department of Clinical Sciences, Lund University, Malmö (MM, OM); Broad Institute of MIT and Harvard, Cambridge, MA (ALS, AAD, KAP, KB, CBC, REG); National Heart, Lung & Blood Institute Division of Intramural Research, Bethesda, MD (CJO); and, Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (TJW)
| | - Clary B Clish
- Framingham Heart Study of the National Heart, Lung and Blood Institute and Boston University School of Medicine, Framingham, MA (SC, MGL, JMM, JEH, CJO, RSV, TJW); Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (SC); Department of Mathematics and Statistics, Boston University, Boston, MA (MGL); Department of Biostatistics, Boston University School of Public Health, Boston, MA (ELM); Cardiology Division (JEH, AG, CJO, REG), Cardiovascular Research Center (REG), and Renal Division (EPR), Massachusetts General Hospital, Harvard Medical School, Boston, MA; General Internal Medicine (JMM), Cardiology (JEH, RSV), and Preventive Medicine (RSV), Department of Medicine, Boston University School of Medicine, Boston, MA; Jean Mayer US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA (PFJ); Department of Clinical Sciences, Lund University, Malmö (MM, OM); Broad Institute of MIT and Harvard, Cambridge, MA (ALS, AAD, KAP, KB, CBC, REG); National Heart, Lung & Blood Institute Division of Intramural Research, Bethesda, MD (CJO); and, Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (TJW)
| | - Ramachandran S Vasan
- Framingham Heart Study of the National Heart, Lung and Blood Institute and Boston University School of Medicine, Framingham, MA (SC, MGL, JMM, JEH, CJO, RSV, TJW); Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (SC); Department of Mathematics and Statistics, Boston University, Boston, MA (MGL); Department of Biostatistics, Boston University School of Public Health, Boston, MA (ELM); Cardiology Division (JEH, AG, CJO, REG), Cardiovascular Research Center (REG), and Renal Division (EPR), Massachusetts General Hospital, Harvard Medical School, Boston, MA; General Internal Medicine (JMM), Cardiology (JEH, RSV), and Preventive Medicine (RSV), Department of Medicine, Boston University School of Medicine, Boston, MA; Jean Mayer US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA (PFJ); Department of Clinical Sciences, Lund University, Malmö (MM, OM); Broad Institute of MIT and Harvard, Cambridge, MA (ALS, AAD, KAP, KB, CBC, REG); National Heart, Lung & Blood Institute Division of Intramural Research, Bethesda, MD (CJO); and, Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (TJW)
| | - Robert E Gerszten
- Framingham Heart Study of the National Heart, Lung and Blood Institute and Boston University School of Medicine, Framingham, MA (SC, MGL, JMM, JEH, CJO, RSV, TJW); Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (SC); Department of Mathematics and Statistics, Boston University, Boston, MA (MGL); Department of Biostatistics, Boston University School of Public Health, Boston, MA (ELM); Cardiology Division (JEH, AG, CJO, REG), Cardiovascular Research Center (REG), and Renal Division (EPR), Massachusetts General Hospital, Harvard Medical School, Boston, MA; General Internal Medicine (JMM), Cardiology (JEH, RSV), and Preventive Medicine (RSV), Department of Medicine, Boston University School of Medicine, Boston, MA; Jean Mayer US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA (PFJ); Department of Clinical Sciences, Lund University, Malmö (MM, OM); Broad Institute of MIT and Harvard, Cambridge, MA (ALS, AAD, KAP, KB, CBC, REG); National Heart, Lung & Blood Institute Division of Intramural Research, Bethesda, MD (CJO); and, Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (TJW)
| | - Thomas J Wang
- Framingham Heart Study of the National Heart, Lung and Blood Institute and Boston University School of Medicine, Framingham, MA (SC, MGL, JMM, JEH, CJO, RSV, TJW); Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (SC); Department of Mathematics and Statistics, Boston University, Boston, MA (MGL); Department of Biostatistics, Boston University School of Public Health, Boston, MA (ELM); Cardiology Division (JEH, AG, CJO, REG), Cardiovascular Research Center (REG), and Renal Division (EPR), Massachusetts General Hospital, Harvard Medical School, Boston, MA; General Internal Medicine (JMM), Cardiology (JEH, RSV), and Preventive Medicine (RSV), Department of Medicine, Boston University School of Medicine, Boston, MA; Jean Mayer US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA (PFJ); Department of Clinical Sciences, Lund University, Malmö (MM, OM); Broad Institute of MIT and Harvard, Cambridge, MA (ALS, AAD, KAP, KB, CBC, REG); National Heart, Lung & Blood Institute Division of Intramural Research, Bethesda, MD (CJO); and, Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (TJW)
| |
Collapse
|
1060
|
Tsuda T. Possible abilities of dietary factors to prevent and treat diabetes via the stimulation of glucagon-like peptide-1 secretion. Mol Nutr Food Res 2015; 59:1264-73. [PMID: 25707985 DOI: 10.1002/mnfr.201400871] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 02/01/2015] [Accepted: 02/09/2015] [Indexed: 12/22/2022]
Abstract
There is a pressing need for countermeasures against diabetes, which has increased in incidence, becoming a global issue. Glucagon-like peptide-1 (GLP-1), a molecule secreted in enteroendocrine L cells in the lower small and large intestines, is thought to be one of the most important molecular targets for the prevention and treatment of diabetes. There has been increasing interest in the possible ability of dietary factors to treat diabetes via modulating GLP-1 secretion. There is thought to be a close relationship between incretin and diet, and the purported best approach for using dietary factors to increase GLP-1 activity is promotion of secretion of endogenous GLP-1. It have been reported that nutrients as well as various non-nutrient dietary factors can function as GLP-1 secretogogues. Here, we present our findings on the GLP-1 secretion-stimulating functions of two dietary factors, curcumin and extract of edible sweet potato leaves, which contain caffeoylquinic acid derivatives. However, it is necessary to reveal in greater detail the stimulation of GLP-1 secretion by dietary factors for preventing and treating diabetes. It is desirable to clarify the exact GLP-1 secretory pathway, the effect of metabolites derived from dietary factors in gut lumen, and the relationship between incretin and meal.
Collapse
Affiliation(s)
- Takanori Tsuda
- College of Bioscience and Biotechnology, Chubu University, Kasugai, Aichi, Japan
| |
Collapse
|
1061
|
Allin KH, Nielsen T, Pedersen O. Mechanisms in endocrinology: Gut microbiota in patients with type 2 diabetes mellitus. Eur J Endocrinol 2015; 172:R167-77. [PMID: 25416725 DOI: 10.1530/eje-14-0874] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Perturbations of the composition and function of the gut microbiota have been associated with metabolic disorders including obesity, insulin resistance and type 2 diabetes. Studies on mice have demonstrated several underlying mechanisms including host signalling through bacterial lipopolysaccharides derived from the outer membranes of Gram-negative bacteria, bacterial fermentation of dietary fibres to short-chain fatty acids and bacterial modulation of bile acids. On top of this, an increased permeability of the intestinal epithelium may lead to increased absorption of macromolecules from the intestinal content resulting in systemic immune responses, low-grade inflammation and altered signalling pathways influencing lipid and glucose metabolism. While mechanistic studies on mice collectively support a causal role of the gut microbiota in metabolic diseases, the majority of studies in humans are correlative of nature and thus hinder causal inferences. Importantly, several factors known to influence the risk of type 2 diabetes, e.g. diet and age, have also been linked to alterations in the gut microbiota complicating the interpretation of correlative studies. However, based upon the available evidence, it is hypothesised that the gut microbiota may mediate or modulate the influence of lifestyle factors triggering development of type 2 diabetes. Thus, the aim of this review is to critically discuss the potential role of the gut microbiota in the pathophysiology and pathogenesis of type 2 diabetes.
Collapse
Affiliation(s)
- Kristine H Allin
- The Novo Nordisk Foundation Center for Basic Metabolic ResearchSection of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 1, DK-2100 Copenhagen Ø, Denmark
| | - Trine Nielsen
- The Novo Nordisk Foundation Center for Basic Metabolic ResearchSection of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 1, DK-2100 Copenhagen Ø, Denmark
| | - Oluf Pedersen
- The Novo Nordisk Foundation Center for Basic Metabolic ResearchSection of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 1, DK-2100 Copenhagen Ø, Denmark
| |
Collapse
|
1062
|
Sombetzki M, Fuchs CD, Fickert P, Österreicher CH, Mueller M, Claudel T, Loebermann M, Engelmann R, Langner C, Sahin E, Schwinge D, Guenther ND, Schramm C, Mueller-Hilke B, Reisinger EC, Trauner M. 24-nor-ursodeoxycholic acid ameliorates inflammatory response and liver fibrosis in a murine model of hepatic schistosomiasis. J Hepatol 2015; 62:871-8. [PMID: 25463533 PMCID: PMC4368108 DOI: 10.1016/j.jhep.2014.11.020] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 11/09/2014] [Accepted: 11/11/2014] [Indexed: 12/21/2022]
Abstract
BACKGROUND & AIMS Intrahepatic granuloma formation and fibrosis characterize the pathological features of Schistosoma mansoni infection. Based on previously observed substantial anti-fibrotic effects of 24-nor-ursodeoxycholic acid (norUDCA) in Abcb4/Mdr2(-/-) mice with cholestatic liver injury and biliary fibrosis, we hypothesized that norUDCA improves inflammation-driven liver fibrosis in S. mansoni infection. METHODS Adult NMRI mice were infected with 50 S. mansoni cercariae and after 12 weeks received either norUDCA- or ursodeoxycholic acid (UDCA)-enriched diet (0.5% wt/wt) for 4 weeks. Bile acid effects on liver histology, serum biochemistry, key regulatory cytokines, hepatic hydroxyproline content as well as granuloma formation were compared to naive mice and infected controls. In addition, effects of norUDCA on primary T-cell activation/proliferation and maturation of the antigen-presenting-cells (dendritic cells, macrophages) were determined in vitro. RESULTS UDCA as well as norUDCA attenuated the inflammatory response in livers of S. mansoni infected mice, but exclusively norUDCA changed cellular composition and reduced size of hepatic granulomas as well as TH2-mediated hepatic fibrosis in vivo. Moreover, norUDCA affected surface expression level of major histocompatibility complex (MHC) class II of macrophages and dendritic cells as well as activation/proliferation of T-lymphocytes in vitro, whereas UDCA had no effect. CONCLUSIONS This study demonstrates pronounced anti-inflammatory and anti-fibrotic effects of norUDCA compared to UDCA in S. mansoni induced liver injury, and indicates that norUDCA directly represses antigen presentation of antigen presenting cells and subsequent T-cell activation in vitro. Therefore, norUDCA represents a promising drug for the treatment of this important cause of liver fibrosis.
Collapse
Affiliation(s)
- Martina Sombetzki
- Division of Tropical Medicine and Infectious Diseases, Department of Internal Medicine, University of Rostock, Germany
| | - Claudia D Fuchs
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Austria
| | - Peter Fickert
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Austria; Institute of Pathology, Medical University Graz, Austria
| | - Christoph H Österreicher
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Austria
| | - Michaela Mueller
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Austria
| | - Thierry Claudel
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Austria
| | - Micha Loebermann
- Division of Tropical Medicine and Infectious Diseases, Department of Internal Medicine, University of Rostock, Germany
| | | | - Cord Langner
- Institute of Pathology, Medical University Graz, Austria
| | - Emine Sahin
- Institute for Physiology, Center for Physiology and Pharmacology, Medical University of Vienna, Austria
| | - Dorothee Schwinge
- Department of Medicine I, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nina D Guenther
- Department of Medicine I, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph Schramm
- Department of Medicine I, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Emil C Reisinger
- Division of Tropical Medicine and Infectious Diseases, Department of Internal Medicine, University of Rostock, Germany
| | - Michael Trauner
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Austria.
| |
Collapse
|
1063
|
Yu DD, Sousa KM, Mattern DL, Wagner J, Fu X, Vaidehi N, Forman BM, Huang W. Stereoselective synthesis, biological evaluation, and modeling of novel bile acid-derived G-protein coupled Bile acid receptor 1 (GP-BAR1, TGR5) agonists. Bioorg Med Chem 2015; 23:1613-28. [DOI: 10.1016/j.bmc.2015.01.048] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 01/19/2015] [Accepted: 01/27/2015] [Indexed: 12/31/2022]
|
1064
|
Kaur A, Patankar JV, de Haan W, Ruddle P, Wijesekara N, Groen AK, Verchere CB, Singaraja RR, Hayden MR. Loss of Cyp8b1 improves glucose homeostasis by increasing GLP-1. Diabetes 2015; 64:1168-79. [PMID: 25338812 DOI: 10.2337/db14-0716] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Besides their role in facilitating lipid absorption, bile acids are increasingly being recognized as signaling molecules that activate cell-signaling receptors. Targeted disruption of the sterol 12α-hydroxylase gene (Cyp8b1) results in complete absence of cholic acid (CA) and its derivatives. Here we investigate the effect of Cyp8b1 deletion on glucose homeostasis. Absence of Cyp8b1 results in improved glucose tolerance, insulin sensitivity, and β-cell function, mediated by absence of CA in Cyp8b1(-/-) mice. In addition, we show that reduced intestinal fat absorption in the absence of biliary CA leads to increased free fatty acids reaching the ileal L cells. This correlates with increased secretion of the incretin hormone GLP-1. GLP-1, in turn, increases the biosynthesis and secretion of insulin from β-cells, leading to the improved glucose tolerance observed in the Cyp8b1(-/-) mice. Thus, our data elucidate the importance of Cyp8b1 inhibition on the regulation of glucose metabolism.
Collapse
Affiliation(s)
- Achint Kaur
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jay V Patankar
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Willeke de Haan
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Piers Ruddle
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Nadeeja Wijesekara
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Albert K Groen
- Departments of Pediatrics and Laboratory Medicine, Center for Liver, Digestive and Metabolic Diseases, University Medical Center Groningen, Groningen, the Netherlands
| | - C Bruce Verchere
- Departments of Surgery and Pathology and Laboratory Medicine, Child & Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Roshni R Singaraja
- A*STAR (Agency for Science, Technology and Research) Institute and Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Michael R Hayden
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
1065
|
Abstract
With the prevalence of cardio-metabolic disorders reaching pandemic proportions, the search for modifiable causative factors has intensified. One such potential factor is the vast microbial community inhabiting the human gastrointestinal tract, the gut microbiota. For the past decade evidence has accumulated showing the association of distinct changes in gut microbiota composition and function with obesity, type 2 diabetes and cardiovascular disease. Although causality in humans and the pathophysiological mechanisms involved have yet to be decisively established, several studies have demonstrated that the gut microbiota, as an environmental factor influencing the metabolic state of the host, is readily modifiable through a variety of interventions. In this review we provide an overview of the development of the gut microbiome and its compositional and functional changes in relation to cardio-metabolic disorders, and give an update on recent progress in how this could be exploited in microbiota-based therapeutics.
Collapse
Affiliation(s)
- Tue H Hansen
- />The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 1, København Ø, 2100 Denmark
| | - Rikke J Gøbel
- />The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 1, København Ø, 2100 Denmark
| | - Torben Hansen
- />The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 1, København Ø, 2100 Denmark
- />Faculty of Health Sciences, University of Southern Denmark, J.B. Winsløws Vej 19-3, Odense C, 5000 Denmark
| | - Oluf Pedersen
- />The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 1, København Ø, 2100 Denmark
| |
Collapse
|
1066
|
Proteomic and metabonomic biomarkers for hepatocellular carcinoma: a comprehensive review. Br J Cancer 2015; 112:1141-56. [PMID: 25826224 PMCID: PMC4385954 DOI: 10.1038/bjc.2015.38] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 11/04/2014] [Accepted: 12/20/2014] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) ranks third in overall global cancer-related mortality. Symptomatic presentation often means advanced disease where potentially curative treatment options become very limited. Numerous international guidelines propose the routine monitoring of those with the highest risk factors for the condition in order to diagnose potential tumourigenesis early. To aid this, the fields of metabonomic- and proteomic-based biomarker discovery have applied advanced tools to identify early changes in protein and metabolite expression in HCC patients vs controls. With robust validation, it is anticipated that from these candidates will rise a high-performance non-invasive test able to diagnose early HCC and related conditions. This review gathers the numerous markers proposed by studies using mass spectrometry and proton nuclear magnetic resonance spectroscopy and evaluates areas of consistency as well as discordance.
Collapse
|
1067
|
Kubitz R, Dröge C, Kluge S, Stindt J, Häussinger D. Genetic variations of bile salt transporters. DRUG DISCOVERY TODAY. TECHNOLOGIES 2015; 12:e55-67. [PMID: 25027376 DOI: 10.1016/j.ddtec.2014.03.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Bile salt transporters directly or indirectly influence biological processes through physicochemical or signalling properties of bile salts. The coordinated action of uptake and efflux transporters in polarized epithelial cells of the liver, biliary tree, small intestine and kidney determine bile salt concentrations in different compartments of the body. Genetic variations of bile salt transporters lead to clinical relevant phenotypes of varying severity ranging from a predisposition for drug-induced liver injury to rapidly progressing end-stage liver disease. This review focuses on the impact of genetic variations of bile salt transporters including BSEP, NTCP, ASBT and OSTα/β and discusses approaches for transporter analysis.
Collapse
|
1068
|
|
1069
|
Cortés V, Amigo L, Zanlungo S, Galgani J, Robledo F, Arrese M, Bozinovic F, Nervi F. Metabolic effects of cholecystectomy: gallbladder ablation increases basal metabolic rate through G-protein coupled bile acid receptor Gpbar1-dependent mechanisms in mice. PLoS One 2015; 10:e0118478. [PMID: 25738495 PMCID: PMC4349594 DOI: 10.1371/journal.pone.0118478] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 01/18/2015] [Indexed: 02/07/2023] Open
Abstract
Background & Aims Bile acids (BAs) regulate energy expenditure by activating G-protein Coupled Bile Acid Receptor Gpbar1/TGR5 by cAMP-dependent mechanisms. Cholecystectomy (XGB) increases BAs recirculation rates resulting in increased tissue exposure to BAs during the light phase of the diurnal cycle in mice. We aimed to determine: 1) the effects of XGB on basal metabolic rate (BMR) and 2) the roles of TGR5 on XGB-dependent changes in BMR. Methods BMR was determined by indirect calorimetry in wild type and Tgr5 deficient (Tgr5-/-) male mice. Bile flow and BAs secretion rates were measured by surgical diversion of biliary duct. Biliary BAs and cholesterol were quantified by enzymatic methods. BAs serum concentration and specific composition was determined by liquid chromatography/tandem mass spectrometry. Gene expression was determined by qPCR analysis. Results XGB increased biliary BAs and cholesterol secretion rates, and elevated serum BAs concentration in wild type and Tgr5-/- mice during the light phase of the diurnal cycle. BMR was ~25% higher in cholecystectomized wild type mice (p <0.02), whereas no changes were detected in cholecystectomized Tgr5-/- mice compared to wild-type animals. Conclusion XGB increases BMR by TGR5-dependent mechanisms in mice.
Collapse
Affiliation(s)
- Víctor Cortés
- Departamento de Nutrición, Diabetes y Metabolismo, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ludwig Amigo
- Departamento de Gastroenterología, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Silvana Zanlungo
- Departamento de Gastroenterología, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - José Galgani
- Departamento de Nutrición, Diabetes y Metabolismo, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fermín Robledo
- Departamento de Gastroenterología, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marco Arrese
- Departamento de Gastroenterología, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Francisco Bozinovic
- Facultad de Medicina, CASEB y Departamento de Ecología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Flavio Nervi
- Departamento de Gastroenterología, Pontificia Universidad Católica de Chile, Santiago, Chile
- * E-mail:
| |
Collapse
|
1070
|
Analysis of the serum bile Acid composition for differential diagnosis in patients with liver disease. Gastroenterol Res Pract 2015; 2015:717431. [PMID: 25821461 PMCID: PMC4363704 DOI: 10.1155/2015/717431] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 02/07/2015] [Accepted: 02/09/2015] [Indexed: 12/14/2022] Open
Abstract
Objectives. We determined the serum bile acid (BA) composition in patients with liver diseases and healthy volunteers to investigate the relationship between the etiologies of liver disease and BA metabolism. Material and Methods. Sera from 150 patients with liver diseases and 46 healthy volunteers were obtained. The serum concentrations of the 16 different BAs were determined according to the LC-MS/MS method and were compared between the different liver diseases. Results. A total of 150 subjects, including patients with hepatitis C virus (HCV) (n = 44), hepatitis B virus (HBV) (n = 23), alcoholic liver disease (ALD) (n = 21), biliary tract disease (n = 20), nonalcoholic fatty liver disease (NAFLD) (n = 13), and other liver diseases (n = 29), were recruited. The levels of UDCA and GUDCA were significantly higher in the ALD group, and the levels of DCA and UDCA were significantly lower in the biliary tract diseases group than in viral hepatitis group. In the UDCA therapy (-) subgroup, a significantly lower level of TLCA was observed in the ALD group, with lower levels of CDCA, DCA, and GLCA noted in biliary tract diseases group compared to viral hepatitis group. Conclusions. Analysis of the BA composition may be useful for differential diagnosis in liver disease.
Collapse
|
1071
|
Ferrebee CB, Dawson PA. Metabolic effects of intestinal absorption and enterohepatic cycling of bile acids. Acta Pharm Sin B 2015; 5:129-34. [PMID: 26579438 PMCID: PMC4629214 DOI: 10.1016/j.apsb.2015.01.001] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 12/30/2014] [Accepted: 01/04/2015] [Indexed: 12/13/2022] Open
Abstract
The classical functions of bile acids include acting as detergents to facilitate the digestion and absorption of nutrients in the gut. In addition, bile acids also act as signaling molecules to regulate glucose homeostasis, lipid metabolism and energy expenditure. The signaling potential of bile acids in compartments such as the systemic circulation is regulated in part by an efficient enterohepatic circulation that functions to conserve and channel the pool of bile acids within the intestinal and hepatobiliary compartments. Changes in hepatobiliary and intestinal bile acid transport can alter the composition, size, and distribution of the bile acid pool. These alterations in turn can have significant effects on bile acid signaling and their downstream metabolic targets. This review discusses recent advances in our understanding of the inter-relationship between the enterohepatic cycling of bile acids and the metabolic consequences of signaling via bile acid-activated receptors, such as farnesoid X nuclear receptor (FXR) and the G-protein-coupled bile acid receptor (TGR5).
Collapse
Key Words
- ACCII, acetyl-CoA carboxylase 2
- APO, apolipoproteins
- ASBT, apical sodium-dependent bile acid transporter
- BSEP, bile salt export pump
- Bile acids
- CYP7A1, cholesterol 7α-hydroxylase
- DIO2, deiodinase 2
- Energy homeostasis
- FAS, fatty acid synthase
- FGF, fibroblast growth factor
- FGFR4, fibroblast growth factor receptor 4
- FOXO1, forkhead box protein O1
- FXR, farnesoid X-receptor
- G6Pase, glucose-6-phosphatase
- GLP-1, glucagon-like polypeptide-1
- HNF4α, hepatocyte nuclear factor 4 alpha
- IBABP, ileal bile acid binding protein
- Intestine
- LDL, low density lipoprotein
- Lipid metabolism
- Liver
- NTCP, Na+-taurocholate transporting polypeptide
- OATP, organic anion transporting polypeptide
- OST, organic solute transporter
- PEPCK, phosphoenolpyruvate carboxykinase
- PGC1α, peroxisome proliferator-activated receptor gamma coactivator 1 alpha
- PPAR, peroxisome proliferator-activated receptor
- SHP, small heterodimer partner
- SREBP1c, sterol regulatory element binding protein-1c
- T4, thyroid hormone
- TGR5, G-protein-coupled bile acid receptor
- Transporters
- VLDL, very low density lipoprotein
Collapse
|
1072
|
Ding L, Yang L, Wang Z, Huang W. Bile acid nuclear receptor FXR and digestive system diseases. Acta Pharm Sin B 2015; 5:135-44. [PMID: 26579439 PMCID: PMC4629217 DOI: 10.1016/j.apsb.2015.01.004] [Citation(s) in RCA: 306] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 12/31/2014] [Accepted: 01/05/2015] [Indexed: 12/14/2022] Open
Abstract
Bile acids (BAs) are not only digestive surfactants but also important cell signaling molecules, which stimulate several signaling pathways to regulate some important biological processes. The bile-acid-activated nuclear receptor, farnesoid X receptor (FXR), plays a pivotal role in regulating bile acid, lipid and glucose homeostasis as well as in regulating the inflammatory responses, barrier function and prevention of bacterial translocation in the intestinal tract. As expected, FXR is involved in the pathophysiology of a wide range of diseases of gastrointestinal tract, including inflammatory bowel disease, colorectal cancer and type 2 diabetes. In this review, we discuss current knowledge of the roles of FXR in physiology of the digestive system and the related diseases. Better understanding of the roles of FXR in digestive system will accelerate the development of FXR ligands/modulators for the treatment of digestive system diseases.
Collapse
Key Words
- 6-ECDCA, 6α-ethyl-chenodeoxycholic acid
- AF2, activation domain
- ANGTPL3, angiopoietin-like protein 3
- AOM, azoxymethane
- AP-1, activator protein-1
- ASBT, apical sodium-dependent bile salt transporter
- Apo, apolipoprotein
- BAAT, bile acid-CoA amino acid N-acetyltransferase
- BACS, bile acid-CoA synthetase
- BAs, bile acids
- BMI, body mass index
- BSEP, bile salt export pump
- Bile acids
- CA, cholic acid
- CD, Crohn׳s disease
- CDCA, chenodeoxycholic acid
- CREB, cAMP regulatory element-binding protein
- CYP7A1, cholesterol 7α-hydroxylase
- Colorectal cancer
- DBD, DNA binding domain
- DCA, deoxycholic acid
- DSS, dextrane sodium sulfate
- ERK, extracellular signal-regulated kinase
- FABP6, fatty acid-binding protein subclass 6
- FFAs, free fatty acids
- FGF19, fibroblast growth factor 19
- FGFR4, fibroblast growth factor receptor 4
- FXR, farnesoid X receptor
- FXRE, farnesoid X receptor response element
- Farnesoid X receptor
- G6Pase, glucose-6-phosphatase
- GLP-1, glucagon-like peptide 1
- GLUT2, glucose transporter type 2
- GPBAR, G protein-coupled BA receptor
- GPCRs, G protein-coupled receptors
- GSK3, glycogen synthase kinase 3
- Gastrointestinal tract
- HDL-C, high density lipoprotein cholesterol
- HNF4α, hepatic nuclear factor 4α
- I-BABP, intestinal bile acid-binding protein
- IBD, inflammatory bowel disease
- IL-1, interleukin 1
- Inflammatory bowel disease
- KLF11, Krüppel-like factor 11
- KRAS, Kirsten rat sarcoma viral oncogene homolog
- LBD, ligand binding domain
- LCA, lithocholic acid
- LPL, lipoprotein lipase
- LRH-1, liver receptor homolog-1
- MCA, muricholicacid
- MRP2, multidrug resistance-associated protein 2
- NF-κB, nuclear factor-kappa B
- NOD, non-obese diabetic
- NRs, nuclear receptors
- OSTα, organic solute transporter alpha
- OSTβ, organic solute transporter beta
- PEPCK, phosphoenol pyruvate carboxykinase
- PGC-1α, peroxisome proliferators-activated receptor γ coactivator protein-1α
- SHP, small heterodimer partner
- SREBP-1c, sterol regulatory element-binding protein 1c
- STAT3, signal transducers and activators of transcription 3
- T2D, type 2 diabetes
- TLCA, taurolithocholic acid
- TNBS, trinitrobenzensulfonic acid
- TNFα, tumor necrosis factors α
- Type 2 diabetes
- UC, ulcerative colitis
- UDCA, ursodeoxycholic acid
- VSG, vertical sleeve gastrectomy
- db/db, diabetic mice
Collapse
|
1073
|
Abstract
PURPOSE OF REVIEW This review focuses on the latest understanding of the molecular mechanisms underlying the complex interactions between intestine and liver bile acid signaling, gut microbiota, and their impact on whole-body lipid, glucose and energy metabolism. RECENT FINDINGS Hepatic bile acid synthesis is tightly regulated by the bile acid negative feedback mechanisms. Modulating the enterohepatic bile acid signaling greatly impacts the whole-body metabolic homeostasis. Recently, a positive feedback mechanism through intestine farnesoid X receptor (FXR) antagonism has been proposed to link gut microbiota to the regulation of bile acid composition and pool size. Two studies identified intestine Diet1 and hepatic SHP-2 as novel regulators of CYP7A1 and bile acid synthesis through the gut-liver FXR-fibroblast growth factor 15/19-FGF receptor four signaling axis. New evidence suggests that enhancing bile acid signaling in the distal ileum and colon contributes to the metabolic benefits of bile acid sequestrants and bariatric surgery. SUMMARY Small-molecule ligands that target TGR5 and FXR have shown promise in treating various metabolic and inflammation-related human diseases. New insights into the mechanisms underlying the bariatric surgery and bile acid sequestrant treatment suggest that targeting the enterohepatic circulation to modulate gut-liver bile acid signaling, incretin production and microbiota represents a new strategy to treat obesity and type 2 diabetes.
Collapse
|
1074
|
Magotti P, Bauer I, Igarashi M, Babagoli M, Marotta R, Piomelli D, Garau G. Structure of human N-acylphosphatidylethanolamine-hydrolyzing phospholipase D: regulation of fatty acid ethanolamide biosynthesis by bile acids. Structure 2015; 23:598-604. [PMID: 25684574 DOI: 10.1016/j.str.2014.12.018] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 12/16/2014] [Accepted: 12/18/2014] [Indexed: 12/31/2022]
Abstract
The fatty acid ethanolamides (FAEs) are lipid mediators present in all organisms and involved in highly conserved biological functions, such as innate immunity, energy balance, and stress control. They are produced from membrane N-acylphosphatidylethanolamines (NAPEs) and include agonists for G protein-coupled receptors (e.g., cannabinoid receptors) and nuclear receptors (e.g., PPAR-α). Here, we report the crystal structure of human NAPE-hydrolyzing phospholipase D (NAPE-PLD) at 2.65 Å resolution, a membrane enzyme that catalyzes FAE formation in mammals. NAPE-PLD forms homodimers partly separated by an internal ∼ 9-Å-wide channel and uniquely adapted to associate with phospholipids. A hydrophobic cavity provides an entryway for NAPE into the active site, where a binuclear Zn(2+) center orchestrates its hydrolysis. Bile acids bind with high affinity to selective pockets in this cavity, enhancing dimer assembly and enabling catalysis. These elements offer multiple targets for the design of small-molecule NAPE-PLD modulators with potential applications in inflammation and metabolic disorders.
Collapse
Affiliation(s)
- Paola Magotti
- Department of Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Inga Bauer
- Department of Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Miki Igarashi
- Department of Anatomy & Neurobiology, University of California - Irvine, Gillespie NRF 3101, Irvine, CA 92697, USA
| | - Masih Babagoli
- Department of Anatomy & Neurobiology, University of California - Irvine, Gillespie NRF 3101, Irvine, CA 92697, USA
| | - Roberto Marotta
- Nanochemistry, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Daniele Piomelli
- Department of Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy; Department of Anatomy & Neurobiology, University of California - Irvine, Gillespie NRF 3101, Irvine, CA 92697, USA.
| | - Gianpiero Garau
- Department of Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy.
| |
Collapse
|
1075
|
Abstract
The enteroendocrine system is the primary sensor of ingested nutrients and is responsible for secreting an array of gut hormones, which modulate multiple physiological responses including gastrointestinal motility and secretion, glucose homeostasis, and appetite. This Review provides an up-to-date synopsis of the molecular mechanisms underlying enteroendocrine nutrient sensing and highlights our current understanding of the neuro-hormonal regulation of gut hormone secretion, including the interaction between the enteroendocrine system and the enteric nervous system. It is hoped that a deeper understanding of how these systems collectively regulate postprandial physiology will further facilitate the development of novel therapeutic strategies.
Collapse
|
1076
|
Svane MS, Bojsen-Møller KN, Madsbad S, Holst JJ. Updates in weight loss surgery and gastrointestinal peptides. Curr Opin Endocrinol Diabetes Obes 2015; 22:21-8. [PMID: 25517025 DOI: 10.1097/med.0000000000000131] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE OF REVIEW Roux-en-Y gastric bypass (RYGB) and sleeve gastrectomy are referred to as 'metabolic surgery' due to hormonal shifts with impacts on diabetes remission and weight loss. The purpose of this review is to summarize recent findings in mechanisms underlying beneficial effects of weight loss surgery. RECENT FINDINGS Importantly, gut hormone secretion is altered after RYGB and sleeve gastrectomy due to accelerated transit of nutrients to distal parts of the small intestine, leading to excessive release of L-cell peptide hormones [e.g. glucagon-like peptide-1 (GLP-1), peptide YY].Improved glucose metabolism after RYGB and sleeve gastrectomy involves several mechanisms: early increased hepatic insulin sensitivity, resulting from reduced liver fat content in response to the postoperative caloric restriction, improved beta-cell function mediated by exaggerated postprandial GLP-1 secretion; as demonstrated by relapse of impaired glucose tolerance in studies blocking the GLP-1 receptor by exendin 9-39, and later after major weight loss increased peripheral insulin sensitivity. Gut hormone secretion changes towards a more anorectic profile and is likely important for less caloric intake and weight loss. SUMMARY Changes in gut hormone secretion after RYGB and sleeve gastrectomy surgery induce the beneficial effects on weight and glycemic control through the influence on appetite regulation and insulin secretion.
Collapse
Affiliation(s)
- Maria Saur Svane
- aDepartment of Endocrinology, Hvidovre University Hospital, Hvidovre bDepartment of Biomedical Sciences and NNF Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | |
Collapse
|
1077
|
Gao S, Ji XF, Li F, Sun FK, Zhao J, Fan YC, Wang K. Aberrant DNA methylation of G-protein-coupled bile acid receptor Gpbar1 predicts prognosis of acute-on-chronic hepatitis B liver failure. J Viral Hepat 2015; 22:112-119. [PMID: 24995843 DOI: 10.1111/jvh.12277] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The G-protein-coupled bile acid receptor Gpbar1 (TGR5) has been demonstrated to be able to negatively regulate hepatic inflammatory response. In this study, we aimed to determine the methylation status of TGR5 promoter in patients with acute-on-chronic hepatitis B liver failure (ACHBLF) and its predictive value for prognosis. We enrolled 76 consecutive ACHBLF patients, 80 chronic hepatitis B (CHB) patients and 30 healthy controls (HCs). Methylation status of TGR5 promoter in peripheral mononuclear cell (PBMC) was detected by methylation-specific polymerase chain reaction (MSP). The mRNA level of TGR5 was determined by quantitative real-time polymerase chain reaction (RT-qPCR). We found that the frequency of TGR5 promoter methylation was significantly higher in ACHBLF (35/76, 46.05%) than CHB patients (5/80, 6.25%; χ(2) = 32.38, P < 0.01) and HCs (1/30, 3.33%; χ(2) = 17.50, P < 0.01). TGR5 mRNA level was significantly lower (Z = -9.12, P < 0.01) in participants with aberrant methylation than those without. TGR5 methylation showed a sensitivity of 46.05% (35/76), specificity of 93.75% (75/80), positive predictive value (PPV) of 87.5% (35/40) and negative predictive value (NPV) of 64.66% (75/116) in discriminating ACHBLF from CHB patients. ACHBLF patients with methylated TGR5 showed significantly poor survival than those without (P < 0.01). When used to predict 3-month mortality of ACHBLF, TGR5 methylation [area under the receiver operating characteristic curve (AUC) = 0.75] performed significantly better than model for end-stage liver diseases (MELD) score (AUC = 0.65; P < 0.05). Therefore, our study demonstrated that aberrant TGR5 promoter methylation occurred in ACHBLF and might be a potential prognostic marker for the disease.
Collapse
Affiliation(s)
- S Gao
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
| | | | | | | | | | | | | |
Collapse
|
1078
|
Martineau MG, Raker C, Dixon PH, Chambers J, Machirori M, King NM, Hooks ML, Manoharan R, Chen K, Powrie R, Williamson C. The metabolic profile of intrahepatic cholestasis of pregnancy is associated with impaired glucose tolerance, dyslipidemia, and increased fetal growth. Diabetes Care 2015; 38:243-8. [PMID: 25504029 DOI: 10.2337/dc14-2143] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Quantification of changes in glucose and lipid concentrations in women with intrahepatic cholestasis of pregnancy (ICP) and uncomplicated pregnancy and study of their influence on fetal growth. RESEARCH DESIGN AND METHODS A prospective study comparing metabolic outcomes in cholestastic and uncomplicated singleton pregnancies was undertaken at two university hospitals in the U.K. and U.S. from 2011-2014. A total of 26 women with ICP and 27 control pregnancies with no prior history of gestational diabetes mellitus were recruited from outpatient antenatal services and followed until delivery. Alterations in glucose, incretins, cholesterol, and triglycerides were studied using a continuous glucose monitoring (CGM) system and/or a standard glucose tolerance test (GTT) in conjunction with GLP-1 and a fasting lipid profile. Fetal growth was quantified using adjusted birth centiles. RESULTS Maternal blood glucose concentrations were significantly increased in ICP during ambulatory CGM (P < 0.005) and following a GTT (P < 0.005). ICP is characterized by increased fasting triglycerides (P < 0.005) and reduced HDL cholesterol (P < 0.005), similar to changes observed in metabolic syndrome. The offspring of mothers with ICP had significantly larger customized birth weight centiles, adjusted for ethnicity, sex, and gestational age (P < 0.005). CONCLUSIONS ICP is associated with impaired glucose tolerance, dyslipidemia, and increased fetal growth. These findings may have implications regarding the future health of affected offspring.
Collapse
Affiliation(s)
- Marcus G Martineau
- Maternal and Fetal Disease Group, Institute of Reproductive and Developmental Biology, Imperial College London, London, U.K. Division of Obstetric and Consultative Medicine, Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI
| | - Christina Raker
- Division of Research, Women & Infants Hospital of Rhode Island, Providence, RI
| | - Peter H Dixon
- Maternal and Fetal Disease Group, Institute of Reproductive and Developmental Biology, Imperial College London, London, U.K. Women's Health Academic Centre, King's College London, London, U.K
| | - Jenny Chambers
- Maternal and Fetal Disease Group, Institute of Reproductive and Developmental Biology, Imperial College London, London, U.K
| | - Mavis Machirori
- Maternal and Fetal Disease Group, Institute of Reproductive and Developmental Biology, Imperial College London, London, U.K
| | - Nicole M King
- Maternal and Fetal Disease Group, Institute of Reproductive and Developmental Biology, Imperial College London, London, U.K
| | - Melissa L Hooks
- Division of Obstetric and Consultative Medicine, Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI
| | - Ramya Manoharan
- Maternal and Fetal Disease Group, Institute of Reproductive and Developmental Biology, Imperial College London, London, U.K
| | - Kenneth Chen
- Division of Obstetric and Consultative Medicine, Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI
| | - Raymond Powrie
- Division of Obstetric and Consultative Medicine, Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI
| | - Catherine Williamson
- Maternal and Fetal Disease Group, Institute of Reproductive and Developmental Biology, Imperial College London, London, U.K. Women's Health Academic Centre, King's College London, London, U.K.
| |
Collapse
|
1079
|
Brønden A, Hansen M, Sonne DP, Rohde U, Vilsbøll T, Knop FK. Sevelamer in a diabetologist's perspective: a phosphate-binding resin with glucose-lowering potential. Diabetes Obes Metab 2015; 17:116-20. [PMID: 25041567 DOI: 10.1111/dom.12355] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 06/30/2014] [Accepted: 07/03/2014] [Indexed: 01/11/2023]
Abstract
Sevelamer is a calcium-free and metal-free phosphate-binding oral drug used in the management of hyperphosphataemia in chronic kidney disease. Preclinical and clinical trials have shown glucose and lipid-lowering effects of sevelamer, thereby giving rise to a potential role of the drug in the treatment of patients with type 2 diabetes. These 'novel' effects are most probably derived from the bile acid-binding properties of sevelamer. The proposed potential is supported by the approval of the bile acid sequestrant colesevelam in the United States for the treatment of type 2 diabetes and hypercholesterolaemia. This article offers a brief review on the effects of sevelamer and a perspective on the potential mechanisms behind the glucose-lowering effect of the drug.
Collapse
Affiliation(s)
- A Brønden
- Department of Medicine, Centre for Diabetes Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; The Danish Diabetes Academy, Odense, Denmark
| | | | | | | | | | | |
Collapse
|
1080
|
Bile acid signaling through farnesoid X and TGR5 receptors in hepatobiliary and intestinal diseases. Hepatobiliary Pancreat Dis Int 2015; 14:18-33. [PMID: 25655287 DOI: 10.1016/s1499-3872(14)60307-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND The well-known functions of bile acids (BAs) are the emulsification and absorption of lipophilic xenobiotics. However, the emerging evidences in the past decade showed that BAs act as signaling molecules that not only autoregulate their own metabolism and enterohepatic recirculation, but also as important regulators of integrative metabolism by activating nuclear and membrane-bound G protein-coupled receptors. The present review was to get insight into the role of maintenance of BA homeostasis and BA signaling pathways in development and management of hepatobiliary and intestinal diseases. DATA SOURCES Detailed and comprehensive search of PubMed and Scopus databases was carried out for original and review articles. RESULTS Disturbances in BA homeostasis contribute to the development of several hepatobiliary and intestinal disorders, such as non-alcoholic fatty liver disease, liver cirrhosis, cholesterol gallstone disease, intestinal diseases and both hepatocellular and colorectal carcinoma. CONCLUSION Further efforts made in order to advance the understanding of sophisticated BA signaling network may be promising in developing novel therapeutic strategies related not only to hepatobiliary and gastrointestinal but also systemic diseases.
Collapse
|
1081
|
Arble DM, Sandoval DA, Seeley RJ. Mechanisms underlying weight loss and metabolic improvements in rodent models of bariatric surgery. Diabetologia 2015; 58:211-20. [PMID: 25374275 PMCID: PMC4289431 DOI: 10.1007/s00125-014-3433-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 10/03/2014] [Indexed: 02/06/2023]
Abstract
Obesity is a growing health risk with few successful treatment options and fewer still that target both obesity and obesity-associated comorbidities. Despite ongoing scientific efforts, the most effective treatment option to date was not developed from basic research but by surgeons observing outcomes in the clinic. Bariatric surgery is the most successful treatment for significant weight loss, resolution of type 2 diabetes and the prevention of future weight gain. Recent work with animal models has shed considerable light on the molecular underpinnings of the potent effects of these 'metabolic' surgical procedures. Here we review data from animal models and how these studies have evolved our understanding of the critical signalling systems that mediate the effects of bariatric surgery. These insights could lead to alternative therapies able to accomplish effects similar to bariatric surgery in a less invasive manner.
Collapse
|
1082
|
Fan M, Wang X, Xu G, Yan Q, Huang W. Bile acid signaling and liver regeneration. BIOCHIMICA ET BIOPHYSICA ACTA 2015; 1849:196-200. [PMID: 24878541 PMCID: PMC4246016 DOI: 10.1016/j.bbagrm.2014.05.021] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Revised: 04/20/2014] [Accepted: 05/20/2014] [Indexed: 12/25/2022]
Abstract
The liver is able to regenerate itself in response to partial hepatectomy or liver injury. This is accomplished by a complex network of different cell types and signals both inside and outside the liver. Bile acids (BAs) are recently identified as liver-specific metabolic signals and promote liver regeneration by activating their receptors: Farnesoid X Receptor (FXR) and G-protein-coupled BA receptor 1 (GPBAR1, or TGR5). FXR is a member of the nuclear hormone receptor superfamily of ligand-activated transcription factors. FXR promotes liver regeneration after 70% partial hepatectomy (PHx) or liver injury. Moreover, activation of FXR is able to alleviate age-related liver regeneration defects. Both liver- and intestine-FXR are activated by BAs after liver resection or injury and promote liver regeneration through distinct mechanism. TGR5 is a membrane-bound BA receptor and it is also activated during liver regeneration. TGR5 regulates BA hydrophobicity and stimulates BA excretion in urine during liver regeneration. BA signaling thus represents a novel metabolic pathway during liver regeneration. This article is part of a Special Issue entitled: Nuclear receptors in animal development.
Collapse
Affiliation(s)
- Mingjie Fan
- Institute of Genetics, College of Life Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Xichun Wang
- Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - Ganyu Xu
- Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - Qingfeng Yan
- Institute of Genetics, College of Life Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Wendong Huang
- Institute of Genetics, College of Life Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China; Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, 1500 E. Duarte Road, Duarte, CA 91010, USA.
| |
Collapse
|
1083
|
Kashihara H, Shimada M, Kurita N, Sato H, Yoshikawa K, Higashijima J, Chikakiyo M, Nishi M, Takasu C. Duodenal-jejunal bypass improves diabetes and liver steatosis via enhanced glucagon-like peptide-1 elicited by bile acids. J Gastroenterol Hepatol 2015; 30:308-15. [PMID: 25088988 DOI: 10.1111/jgh.12690] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/18/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIM Bariatric surgery not only elicits weight loss but also rapidly resolves diabetes. However, the mechanisms remain unclear. The present study investigates how diabetes and liver steatosis are improved after duodenal-jejunal bypass (DJB) compared with a glucagon-like peptide-1 (GLP-1) analog and correlations between bile acids and GLP-1 secretion. METHODS We initially determined the effects of bile acids on GLP-1 in vitro and then assigned 12 male 16-week-old Otsuka Long-Evans Tokushima Fatty rats to groups that underwent DJB, a sham operation, or were treated with the GLP-1 receptor agonist, liraglutide (n = 4 each). Blood glucose, insulin, GLP-1, serum bile acids, liver steatosis, and the number of GLP-1 positive cells (L cells) in the small intestine and colon were investigated in the three groups at eight weeks postoperatively. RESULTS Levels of GLP-1mRNA were upregulated and GLP-1 secretion increased in cells incubated with bile acids in vitro. Weight gain was suppressed more in the DJB than in the sham group in vivo. Diabetes was more improved and GLP-1 levels were significantly higher in the DJB than in the sham group. Serum bile acids were significantly increased, the number of L cells in the ileum was upregulated compared with the sham group, and liver steatosis was significantly improved in the DJB compared with the other two groups. CONCLUSIONS Duodenal-jejunal bypass might improve diabetes and liver steatosis by enhancing GLP-1 secretion through increasing serum bile acids and the proliferation of L cells in the ileum, compared with liraglutide.
Collapse
Affiliation(s)
- Hideya Kashihara
- Department of Surgery, Institute of Health Biosciences, University of Tokushima, Tokushima, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
1084
|
Abstract
A plasma membrane-bound G protein-coupled receptor, TGR5, that transmits bile acid signaling into a cellular response primarily via the cAMP pathway is expressed in human and rodent cholangiocytes and is localized to multiple, diverse subcellular compartments, including primary cilia. Ciliary-associated TGR5 plays an important role in cholangiocyte physiology and may contribute to a group of liver diseases referred to as the 'cholangiociliopathies', which include polycystic liver disease (PLD) and, possibly, cholangiocarcinoma and primary sclerosing cholangitis. Based on our observations that (1) ciliated and nonciliated cholangiocytes respond to TGR5 activation differently (i.e. the level of cAMP increases in nonciliated cholangiocytes but decreases in ciliated cells) and (2) hepatic cysts are derived from cholangiocytes that are characterized by both malformed cilia and increased cAMP levels, we hypothesized that TGR5-mediated cAMP signaling in cystic cholangiocytes contributes to hepatic cystogenesis. Indeed, our studies show that TGR5 is overexpressed and mislocalized in cystic cholangiocytes, and when activated by ligands, results in increased intracellular cAMP levels, cholangiocyte hyperproliferation and cyst growth. Our studies also show that genetic elimination of TGR5 in an animal model of PLD inhibits hepatic cystogenesis. Collectively, these data suggest the involvement of TGR5 in PLD and that TGR5 targeting in cystic cholangiocytes may have therapeutic potential.
Collapse
Affiliation(s)
- Tatyana V Masyuk
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic College of Medicine, Rochester, Minn., USA
| | | | | |
Collapse
|
1085
|
Aw W, Fukuda S. The Role of Integrated Omics in Elucidating the Gut Microbiota Health Potentials. ACTA ACUST UNITED AC 2015. [DOI: 10.1007/978-3-319-23213-3_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
1086
|
|
1087
|
Abstract
The human gastrointestinal tract is home to trillions of bacteria, which vastly outnumber host cells in the body. Although generally overlooked in the field of endocrinology, gut microbial symbionts organize to form a key endocrine organ that converts nutritional cues from the environment into hormone-like signals that impact both normal physiology and chronic disease in the human host. Recent evidence suggests that several gut microbial-derived products are sensed by dedicated host receptor systems to alter cardiovascular disease (CVD) progression. In fact, gut microbial metabolism of dietary components results in the production of proatherogenic circulating factors that act through a meta-organismal endocrine axis to impact CVD risk. Whether pharmacological interventions at the level of the gut microbial endocrine organ will reduce CVD risk is a key new question in the field of cardiovascular medicine. Here we discuss the opportunities and challenges that lie ahead in targeting meta-organismal endocrinology for CVD prevention.
Collapse
Affiliation(s)
- J. Mark Brown
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Stanley L. Hazen
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| |
Collapse
|
1088
|
Lin M, Xie Z, Zhou Y, Li Y, Ren J, Peng XX, Yao M, Yang Z, Liao Q. Dynamic metabonomic and microbiological response of rats to lincomycin exposure: an integrated microbiology and metabonomics analysis. RSC Adv 2015. [DOI: 10.1039/c5ra10626e] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
We investigated the changes of gut microbiome, host metabolism and their relationships of lincomycin exposure by microbiological and metabolomics profiling.
Collapse
Affiliation(s)
- Manna Lin
- School of Chinese Materia Medica
- Guangzhou University of Chinese Medicine
- Guangzhou
- P. R. China
- School of Pharmaceutical Sciences
| | - Zhiyong Xie
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou
- P. R. China
| | - Yuting Zhou
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou
- P. R. China
| | - Yemeng Li
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou
- P. R. China
| | - Jian Ren
- School of Life Sciences
- Sun Yat-sen University
- Guangzhou
- P. R. China
| | - Xuan-xian Peng
- School of Life Sciences
- Sun Yat-sen University
- Guangzhou
- P. R. China
| | - Meicun Yao
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou
- P. R. China
| | - Zhongzhou Yang
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou
- P. R. China
| | - Qiongfeng Liao
- School of Chinese Materia Medica
- Guangzhou University of Chinese Medicine
- Guangzhou
- P. R. China
| |
Collapse
|
1089
|
Jurowich CF, Otto C, Rikkala PR, Wagner N, Vrhovac I, Sabolić I, Germer CT, Koepsell H. Ileal Interposition in Rats with Experimental Type 2 Like Diabetes Improves Glycemic Control Independently of Glucose Absorption. J Diabetes Res 2015; 2015:490365. [PMID: 26185767 PMCID: PMC4491588 DOI: 10.1155/2015/490365] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 06/05/2015] [Accepted: 06/07/2015] [Indexed: 02/08/2023] Open
Abstract
Bariatric operations in obese patients with type 2 diabetes often improve diabetes before weight loss is observed. In patients mainly Roux-en-Y-gastric bypass with partial stomach resection is performed. Duodenojejunal bypass (DJB) and ileal interposition (IIP) are employed in animal experiments. Due to increased glucose exposition of L-cells located in distal ileum, all bariatric surgery procedures lead to higher secretion of antidiabetic glucagon like peptide-1 (GLP-1) after glucose gavage. After DJB also downregulation of Na(+)-d-glucose cotransporter SGLT1 was observed. This suggested a direct contribution of decreased glucose absorption to the antidiabetic effect of bariatric surgery. To investigate whether glucose absorption is also decreased after IIP, we induced diabetes with decreased glucose tolerance and insulin sensitivity in male rats and investigated effects of IIP on diabetes and SGLT1. After IIP, we observed weight-independent improvement of glucose tolerance, increased insulin sensitivity, and increased plasma GLP-1 after glucose gavage. The interposed ileum was increased in diameter and showed increased length of villi, hyperplasia of the epithelial layer, and increased number of L-cells. The amount of SGLT1-mediated glucose uptake in interposed ileum was increased 2-fold reaching the same level as in jejunum. Thus, improvement of glycemic control by bariatric surgery does not require decreased glucose absorption.
Collapse
Affiliation(s)
- Christian Ferdinand Jurowich
- Department of General, Visceral, Vascular and Paediatric Surgery, University Hospital of Würzburg, Oberdürrbacher Straße 6, 97080 Würzburg, Germany
| | - Christoph Otto
- Department of General, Visceral, Vascular and Paediatric Surgery, University Hospital of Würzburg, Oberdürrbacher Straße 6, 97080 Würzburg, Germany
| | - Prashanth Reddy Rikkala
- Institute of Anatomy and Cell Biology, University of Würzburg, Koellikerstraße 6, 97070 Würzburg, Germany
| | - Nicole Wagner
- Institute of Anatomy and Cell Biology, University of Würzburg, Koellikerstraße 6, 97070 Würzburg, Germany
| | - Ivana Vrhovac
- Molecular Toxicology Unit, Institute for Medical Research & Occupational Health, Ksaverska Cesta 2, 10000 Zagreb, Croatia
| | - Ivan Sabolić
- Molecular Toxicology Unit, Institute for Medical Research & Occupational Health, Ksaverska Cesta 2, 10000 Zagreb, Croatia
| | - Christoph-Thomas Germer
- Department of General, Visceral, Vascular and Paediatric Surgery, University Hospital of Würzburg, Oberdürrbacher Straße 6, 97080 Würzburg, Germany
| | - Hermann Koepsell
- Institute of Anatomy and Cell Biology, University of Würzburg, Koellikerstraße 6, 97070 Würzburg, Germany
- Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute, University of Würzburg, Julius-von-Sachs-Platz 2, 97082 Würzburg, Germany
- *Hermann Koepsell:
| |
Collapse
|
1090
|
Martoni CJ, Labbé A, Ganopolsky JG, Prakash S, Jones ML. Changes in bile acids, FGF-19 and sterol absorption in response to bile salt hydrolase active L. reuteri NCIMB 30242. Gut Microbes 2015; 6:57-65. [PMID: 25612224 PMCID: PMC4615650 DOI: 10.1080/19490976.2015.1005474] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 12/03/2014] [Accepted: 01/05/2015] [Indexed: 02/07/2023] Open
Abstract
The size and composition of the circulating bile acid (BA) pool are important factors in regulating the human gut microbiota. Disrupted regulation of BA metabolism is implicated in several chronic diseases. Bile salt hydrolase (BSH)-active Lactobacillus reuteri NCIMB 30242, previously shown to decrease LDL-cholesterol and increase circulating BA, was investigated for its dose response effect on BA profile in a pilot clinical study. Ten otherwise healthy hypercholesterolemic adults, recruited from a clinical trial site in London, ON, were randomized to consume delayed release or standard release capsules containing L. reuteri NCIMB 30242 in escalating dose over 4 weeks. In another aspect, 4 healthy normocholesterolemic subjects with LDL-C below 3.4 mmol/l received delayed release L. reuteri NCIMB 30242 at a constant dose over 4 weeks. The primary outcome measure was the change in plasma BA profile over the intervention period. Additional outcomes included circulating fibroblast growth factor (FGF)-19, plant sterols and LDL-cholesterol as well as fecal microbiota and bsh gene presence. After one week of intervention subjects receiving delayed release L. reuteri NCIMB 30242 increased total BA by 1.13 ± 0.67 μmol/l (P = 0.02), conjugated BA by 0.67 ± 0.39 μmol/l (P = 0.02) and unconjugated BA by 0.46 ± 0.43 μmol/l (P = 0.07), which represented a greater than 2-fold change relative to baseline. Increases in BA were largely maintained post-week 1 and were generally correlated with FGF-19 and inversely correlated with plant sterols. This is the first clinical support showing that a BSH-active probiotic can significantly and rapidly influence BA metabolism and may prove useful in chronic diseases beyond hypercholesterolemia.
Collapse
Affiliation(s)
| | | | | | - Satya Prakash
- Biomedical Technology and Cell Therapy Research Laboratory; Dept. of Biomedical Engineering; Faculty of Medicine; McGill University; Montreal, QC Canada
- Micropharma Limited; Montreal, QC Canada
| | - Mitchell L Jones
- Biomedical Technology and Cell Therapy Research Laboratory; Dept. of Biomedical Engineering; Faculty of Medicine; McGill University; Montreal, QC Canada
- Micropharma Limited; Montreal, QC Canada
| |
Collapse
|
1091
|
Han JL, Lin HL. Intestinal microbiota and type 2 diabetes: From mechanism insights to therapeutic perspective. World J Gastroenterol 2014; 20:17737-17745. [PMID: 25548472 PMCID: PMC4273124 DOI: 10.3748/wjg.v20.i47.17737] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Revised: 09/26/2014] [Accepted: 11/19/2014] [Indexed: 02/06/2023] Open
Abstract
The incidence of type 2 diabetes (T2DM) is rapidly increasing worldwide. However, the pathogenesis of T2DM has not yet been well explained. Recent evidence suggests that the intestinal microbiota composition is associated with obesity and T2DM. In this review, we provide an overview about the mechanisms underlying the role of intestinal microbiota in the pathogenesis of T2DM. There is clear evidence that the intestinal microbiota influences the host through its effect on body weight, bile acid metabolism, proinflammatory activity and insulin resistance, and modulation of gut hormones. Modulating gut microbiota with the use of probiotics, prebiotics, antibiotics, and fecal microbiota transplantation may have benefits for improvement in glucose metabolism and insulin resistance in the host. Further studies are required to increase our understanding of the complex interplay between intestinal microbiota and the host with T2DM. Further studies may be able to boost the development of new effective therapeutic approaches for T2DM.
Collapse
|
1092
|
Jacinto S, Fang S. Essential roles of bile acid receptors FXR and TGR5 as metabolic regulators. Anim Cells Syst (Seoul) 2014. [DOI: 10.1080/19768354.2014.987318] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
1093
|
Labbé A, Ganopolsky JG, Martoni CJ, Prakash S, Jones ML. Bacterial bile metabolising gene abundance in Crohn's, ulcerative colitis and type 2 diabetes metagenomes. PLoS One 2014; 9:e115175. [PMID: 25517115 PMCID: PMC4269443 DOI: 10.1371/journal.pone.0115175] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 11/19/2014] [Indexed: 12/17/2022] Open
Abstract
We performed an analysis to determine the importance of bile acid modification genes in the gut microbiome of inflammatory bowel disease and type 2 diabetic patients. We used publicly available metagenomic datasets from the Human Microbiome Project and the MetaHIT consortium, and determined the abundance of bile salt hydrolase gene (bsh), 7 alpha-dehydroxylase gene (adh) and 7-alpha hydroxysteroid dehydrogenase gene (hsdh) in fecal bacteria in diseased populations of Crohn's disease (CD), Ulcerative Colitis (UC) and Type 2 diabetes mellitus (T2DM). Phylum level abundance analysis showed a significant reduction in Firmicute-derived bsh in UC and T2DM patients but not in CD patients, relative to healthy controls. Reduction of adh and hsdh genes was also seen in UC and T2DM patients, while an increase was observed in the CD population as compared to healthy controls. A further analysis of the bsh genes showed significant differences in the correlations of certain Firmicutes families with disease or healthy populations. From this observation we proceeded to analyse BSH protein sequences and identified BSH proteins clusters representing the most abundant strains in our analysis of Firmicute bsh genes. The abundance of the bsh genes corresponding to one of these protein clusters was significantly reduced in all disease states relative to healthy controls. This cluster includes bsh genes derived from Lachospiraceae, Clostridiaceae, Erysipelotrichaceae and Ruminococcaceae families. This metagenomic analysis provides evidence of the importance of bile acid modifying enzymes in health and disease. It further highlights the importance of identifying gene and protein clusters, as the same gene may be associated with health or disease, depending on the strains expressing the enzyme, and differences in the enzymes themselves.
Collapse
Affiliation(s)
- Alain Labbé
- Micropharma Limited, Montreal, Quebec, Canada
| | | | | | - Satya Prakash
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, Montreal, Quebec, Canada
- Micropharma Limited, Montreal, Quebec, Canada
| | - Mitchell L. Jones
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, Montreal, Quebec, Canada
- Micropharma Limited, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
1094
|
Lieu T, Jayaweera G, Zhao P, Poole DP, Jensen D, Grace M, McIntyre P, Bron R, Wilson YM, Krappitz M, Haerteis S, Korbmacher C, Steinhoff MS, Nassini R, Materazzi S, Geppetti P, Corvera CU, Bunnett NW. The bile acid receptor TGR5 activates the TRPA1 channel to induce itch in mice. Gastroenterology 2014; 147:1417-28. [PMID: 25194674 PMCID: PMC4821165 DOI: 10.1053/j.gastro.2014.08.042] [Citation(s) in RCA: 168] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 08/19/2014] [Accepted: 08/26/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Patients with cholestatic disease have increased systemic concentrations of bile acids (BAs) and profound pruritus. The G-protein-coupled BA receptor 1 TGR5 (encoded by GPBAR1) is expressed by primary sensory neurons; its activation induces neuronal hyperexcitability and scratching by unknown mechanisms. We investigated whether the transient receptor potential ankyrin 1 (TRPA1) is involved in BA-evoked, TGR5-dependent pruritus in mice. METHODS Co-expression of TGR5 and TRPA1 in cutaneous afferent neurons isolated from mice was analyzed by immunofluorescence, in situ hybridization, and single-cell polymerase chain reaction. TGR5-induced activation of TRPA1 was studied in in HEK293 cells, Xenopus laevis oocytes, and primary sensory neurons by measuring Ca(2+) signals. The contribution of TRPA1 to TGR5-induced release of pruritogenic neuropeptides, activation of spinal neurons, and scratching behavior were studied using TRPA1 antagonists or Trpa1(-/-) mice. RESULTS TGR5 and TRPA1 protein and messenger RNA were expressed by cutaneous afferent neurons. In HEK cells, oocytes, and neurons co-expressing TGR5 and TRPA1, BAs caused TGR5-dependent activation and sensitization of TRPA1 by mechanisms that required Gβγ, protein kinase C, and Ca(2+). Antagonists or deletion of TRPA1 prevented BA-stimulated release of the pruritogenic neuropeptides gastrin-releasing peptide and atrial natriuretic peptide B in the spinal cord. Disruption of Trpa1 in mice blocked BA-induced expression of Fos in spinal neurons and prevented BA-stimulated scratching. Spontaneous scratching was exacerbated in transgenic mice that overexpressed TRG5. Administration of a TRPA1 antagonist or the BA sequestrant colestipol, which lowered circulating levels of BAs, prevented exacerbated spontaneous scratching in TGR5 overexpressing mice. CONCLUSIONS BAs induce pruritus in mice by co-activation of TGR5 and TRPA1. Antagonists of TGR5 and TRPA1, or inhibitors of the signaling mechanism by which TGR5 activates TRPA1, might be developed for treatment of cholestatic pruritus.
Collapse
Affiliation(s)
- TinaMarie Lieu
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia
| | - Gihan Jayaweera
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia
| | - Peishen Zhao
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia
| | - Daniel P. Poole
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia,Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| | - Dane Jensen
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia
| | - Megan Grace
- Health Innovations Research Institute and School of Medical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Peter McIntyre
- Health Innovations Research Institute and School of Medical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Romke Bron
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| | - Yvette M. Wilson
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| | - Matteus Krappitz
- Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Silke Haerteis
- Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christoph Korbmacher
- Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Martin S. Steinhoff
- Charles Institute for Translational Dermatology, University College Dublin, Dublin, Ireland
| | - Romina Nassini
- Department of Health Sciences, Clinical Pharmacology Unit, University of Florence, Florence, Italy
| | - Serena Materazzi
- Department of Health Sciences, Clinical Pharmacology Unit, University of Florence, Florence, Italy
| | - Pierangelo Geppetti
- Department of Health Sciences, Clinical Pharmacology Unit, University of Florence, Florence, Italy
| | - Carlos U. Corvera
- Department of Surgery, University of California, San Francisco, San Francisco, California
| | - Nigel W. Bunnett
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia,Department of Pharmacology, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
1095
|
Jiang CM, Pu CW, Hou YH, Chen Z, Alanazy M, Hebbard L. Non alcoholic steatohepatitis a precursor for hepatocellular carcinoma development. World J Gastroenterol 2014; 20:16464-16473. [PMID: 25469014 PMCID: PMC4248189 DOI: 10.3748/wjg.v20.i44.16464] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 06/24/2014] [Accepted: 08/28/2014] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is increasing in prevalence and is one of the most common cancers in the world. Chief amongst the risks of attaining HCC are hepatitis B and C infection, aflatoxin B1 ingestion, alcoholism and obesity. The later has been shown to promote non alcoholic fatty liver disease, which can lead to the inflammatory form non alcoholic steatohepatitis (NASH). NASH is a complex metabolic disorder that can impact greatly on hepatic function. The mechanisms by which NASH promotes HCC are only beginning to be characterized. Here in this review, we give an overview of the recent novel mechanisms published that have been associated with NASH and subsequent HCC progression. We will focus our discussion on inflammation and gut derived inflammation and how they contribute to NASH driven HCC.
Collapse
|
1096
|
Arslan N. Obesity, fatty liver disease and intestinal microbiota. World J Gastroenterol 2014; 20:16452-16463. [PMID: 25469013 PMCID: PMC4248188 DOI: 10.3748/wjg.v20.i44.16452] [Citation(s) in RCA: 137] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Revised: 05/14/2014] [Accepted: 08/13/2014] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a chronic liver disorder that is increasing in prevalence with the worldwide epidemic of obesity. NAFLD is the hepatic manifestation of the metabolic syndrome. The term NAFLD describes a spectrum of liver pathology ranges from simple steatosis to steatosis with inflammation nonalcoholic steatohepatitis and even cirrhosis. Metabolic syndrome and NAFLD also predict hepatocellular carcinoma. Many genetic and environmental factors have been suggested to contribute to the development of obesity and NAFLD, but the exact mechanisms are not known. Intestinal ecosystem contains trillions of microorganisms including bacteria, Archaea, yeasts and viruses. Several studies support the relationship between the intestinal microbial changes and obesity and also its complications, including insulin resistance and NAFLD. Given that the gut and liver are connected by the portal venous system, it makes the liver more vulnerable to translocation of bacteria, bacterial products, endotoxins or secreted cytokines. Altered intestinal microbiota (dysbiosis) may stimulate hepatic fat deposition through several mechanisms: regulation of gut permeability, increasing low-grade inflammation, modulation of dietary choline metabolism, regulation of bile acid metabolism and producing endogenous ethanol. Regulation of intestinal microbial ecosystem by diet modifications or by using probiotics and prebiotics as a treatment for obesity and its complications might be the issue of further investigations.
Collapse
|
1097
|
Mobraten K, Haugbro T, Karlstrom E, Kleiveland CR, Lea T. Activation of the bile acid receptor TGR5 enhances LPS-induced inflammatory responses in a human monocytic cell line. J Recept Signal Transduct Res 2014; 35:402-9. [PMID: 25418122 DOI: 10.3109/10799893.2014.986744] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Bile acids are recognized as signaling molecules, mediating their effects both through the cell surface receptor TGR5 and the nuclear receptor FXR. After a meal, approximately 95% of the bile acids are transported from terminal ileum and back to the liver via the portal vein, resulting in postprandial elevations of bile acids in blood. During the digestion of fat, components from the microbiota, including LPS, are thought to reach the circulation where it may lead to inflammatory responses after binding TLR4 immune cells. Both LPS and bile acids are present in blood after a high-fat meal; we therefore wanted to study consequences of a possible interplay between TGR5 and TLR4 in human monocytes. METHODS The monocytic cell line U937 stably transfected with the NF-κB reporter plasmid 3x-κB-luc was used as a model system to study the effects of TGR5 and TLR4. Activation of MAP kinases was studied to reveal functional consequences of triggering TGR5 in U937 cells. Effects of TGR5 and TLR4 activation were monitored using NF-κB luciferase assay and by quantification of the pro-inflammatory cytokines IL-6 and IL-8 using ELISA. RESULTS In this study, results show that triggering TGR5 with the specific agonist betulinic acid (BA), and the bile acids CDCA or DCA, activated both the main MAP kinases ERK1/2, p38 and JNK, and the NF-κB signaling pathway. We further demonstrated that co-triggering of TLR4 and TGR5 enhanced the activation of NF-κB and the release of inflammatory cytokines in a synergistic manner compared to triggering of TLR4 alone. CONCLUSIONS Thus, two different and simultaneous events associated with the digestive process coordinately affect the function of human monocytes and contribute to enhanced inflammation. Because elevated levels of circulatory LPS may contribute to the development of insulin resistance, the results from this study suggest that bile acids through the activation of TGR5 may have a role in the development of insulin resistance as well.
Collapse
Affiliation(s)
- Kaia Mobraten
- a Department of Chemistry , Biotechnology and Food Science, Norwegian University of Life Sciences , Ås , Norway and
| | - Tarjei Haugbro
- a Department of Chemistry , Biotechnology and Food Science, Norwegian University of Life Sciences , Ås , Norway and
| | - Ellen Karlstrom
- a Department of Chemistry , Biotechnology and Food Science, Norwegian University of Life Sciences , Ås , Norway and
| | - Charlotte R Kleiveland
- a Department of Chemistry , Biotechnology and Food Science, Norwegian University of Life Sciences , Ås , Norway and.,b Quality and Research Department , Ostfold Hospital Trust , Fredrikstad , Norway
| | - Tor Lea
- a Department of Chemistry , Biotechnology and Food Science, Norwegian University of Life Sciences , Ås , Norway and
| |
Collapse
|
1098
|
Abstract
The gut microbiota has emerged as an integral factor that impacts host metabolism and has been suggested to play a vital role in metabolic diseases such as obesity, insulin resistance, type 2 diabetes, and cardiovascular disease. In humans, cross-sectional studies have identified microbiota profiles associated with metabolic diseases, whereas causation mainly has been demonstrated in animal models. Recent studies involving microbiota-based interventions in humans, or transfer of disease-associated microbiota into germ-free mice, underscore that an altered microbiota may directly modulate host metabolism in humans. However, it will be essential to determine whether an altered gut microbiota precedes development of insulin resistance and diabetes and to identify the underlying molecular mechanisms. Increased mechanistic insights of how the microbiota modulates metabolic disease in humans may pave the way for identification of innovative microbiota-based diagnostics and/or therapeutics.
Collapse
|
1099
|
Perino A, Pols TWH, Nomura M, Stein S, Pellicciari R, Schoonjans K. TGR5 reduces macrophage migration through mTOR-induced C/EBPβ differential translation. J Clin Invest 2014; 124:5424-36. [PMID: 25365223 DOI: 10.1172/jci76289] [Citation(s) in RCA: 166] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 09/30/2014] [Indexed: 02/06/2023] Open
Abstract
The bile acid-responsive G protein-coupled receptor TGR5 is involved in several metabolic processes, and recent studies suggest that TGR5 activation may promote pathways that are protective against diet-induced diabetes. Here, we investigated the role of macrophage-specific TGR5 signaling in protecting adipose tissue from inflammation and associated insulin resistance. Examination of adipose tissue from obese mice lacking macrophage Tgr5 revealed enhanced inflammation, increased chemokine expression, and higher macrophage numbers compared with control obese animals. Moreover, macrophage-specific deletion of Tgr5 exacerbated insulin resistance in obese animals. Conversely, pharmacological activation of TGR5 markedly decreased LPS-induced chemokine expression in primary macrophages. This reduction was mediated by AKT-dependent activation of mTOR complex 1, which in turn induced the differential translation of the dominant-negative C/EBPβ isoform, liver inhibitory protein (LIP). Overall, these studies reveal a signaling pathway downstream of TGR5 that modulates chemokine expression in response to high-fat diet and suggest that targeting this pathway has the potential to be therapeutically exploited for prevention of chronic inflammatory diseases and type 2 diabetes mellitus.
Collapse
|
1100
|
Wang XY, Zhang SY, Li J, Liu HN, Xie X, Nan FJ. Highly lipophilic 3-epi-betulinic acid derivatives as potent and selective TGR5 agonists with improved cellular efficacy. Acta Pharmacol Sin 2014; 35:1463-72. [PMID: 25283506 DOI: 10.1038/aps.2014.97] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 05/17/2014] [Indexed: 12/31/2022]
Abstract
AIM TGR5 is a G protein-coupled receptor that is expressed in intestinal L-cells and stimulates glucagon-like peptide 1 (GLP-1) secretion. TGR5 may represent a novel target for the treatment of metabolic disorder. Here, we sought to design and synthesize a series of TGR5 agonists derived from the natural product betulinic acid. METHODS A series of betulinic acid derivatives were designed and synthesized. A cAMP assay was established using a HEK293 cell line expressing human TGR5. Luciferase reporter assay was established using HEK293 cells transfected with plasmids encoding human FXR and luciferase reporter. A human intestinal L-cell line NCI-H716 was used to evaluate the effects of the betulinic acid derivatives on GLP-1 secretion in vitro. RESULTS Biological data revealed that the 3-α-OH triterpenoids consistently show increased potency for TGR5 compared to their 3-β-OH epimers. 3-OH esterification increased the lipophilicity and TGR5 activity of 3-α betulinic derivatives and enhanced the activity differences between 3-α and 3-β derivatives. The 3-α-acyloxy betulinic acids also exhibited a significant dose-dependent GLP-1 secretion effect. CONCLUSION This study demonstrates that highly lipophilic 3-epi-betulinic acid derivatives can be potent and selective TGR5 agonists with improved cellular efficacy, and our research here provides a new strategy for the design and development of potent TGR5 agonists.
Collapse
|