1101
|
Kim K, Lee YM, Rhyu MR, Kim HY. Spergularia marina induces glucagon-like peptide-1 secretion in NCI-H716 cells through bile acid receptor activation. J Med Food 2014; 17:1197-203. [PMID: 25260089 PMCID: PMC4224038 DOI: 10.1089/jmf.2013.3091] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 08/25/2014] [Indexed: 12/11/2022] Open
Abstract
Spergularia marina Griseb. (SM) is a halophyte that grows in mud flats. The aerial portions of SM have been eaten as vegetables and traditionally used to prevent chronic diseases in Korea. However, there has been no scientific report that demonstrates the pharmacological effects of SM. Glucagon-like peptide-1 (GLP-1) is important for the maintenance of glucose and energy homeostasis through acting as a signal in peripheral and neural systems. To discover a functional food for regulating glucose and energy homeostasis, we evaluated the effect of an aqueous ethanolic extract (AEE) of SM on GLP-1 release from enteroendocrine NCI-H716 cells. In addition, we explored the Takeda G-protein-coupled receptor 5 (TGR5) agonist activity of AEE-SM in Chinese hamster ovary (CHO)-K1 cells transiently transfected with human TGR5. As a result, treatment of NCI-H716 cells with AEE-SM increased GLP-1 secretion and intracellular Ca(2+) and cyclic AMP (cAMP) levels in a dose-dependent manner. Transfection of NCI-H716 cells with TGR5-specific small interference RNA inhibited AEE-SM-induced GLP-1 secretion and the increase in Ca(2+) and cAMP levels. Moreover, AEE-SM showed that the TGR5 agonist activity in CHO-K1 cells transiently transfected with TGR5. The results suggest that AEE-SM might be a candidate for a functional food to regulate glucose and energy homeostasis.
Collapse
Affiliation(s)
- Kyong Kim
- Research Group of Food Functionality, Division of Creative Food Science for Health, Korea Food Research Institute , Sungnam-si, Korea
| | | | | | | |
Collapse
|
1102
|
Novel opportunities for next-generation probiotics targeting metabolic syndrome. Curr Opin Biotechnol 2014; 32:21-27. [PMID: 25448228 DOI: 10.1016/j.copbio.2014.10.006] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 09/30/2014] [Accepted: 10/15/2014] [Indexed: 02/07/2023]
Abstract
Various studies have described the beneficial effects of specific bacteria on the characteristics of metabolic syndrome. Intestinal microbiota might therefore represent a modifiable trait for translational intervention to improve the metabolic profiles of obese and type 2 diabetic patients. However, identifying potential probiotic strains that can effectively colonize the gastrointestinal tract and significantly affect host metabolism has been challenging. This review aims to summarize the notable advances and contributions in the field that may prove useful for identifying next-generation probiotics that target metabolic syndrome and its related disorders.
Collapse
|
1103
|
Lieu T, Jayaweera G, Bunnett NW. GPBA: a GPCR for bile acids and an emerging therapeutic target for disorders of digestion and sensation. Br J Pharmacol 2014; 171:1156-66. [PMID: 24111923 DOI: 10.1111/bph.12426] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 09/03/2013] [Accepted: 09/16/2013] [Indexed: 01/04/2023] Open
Abstract
Bile acids (BAs) are digestive secretions that are necessary for the emulsification and absorption of dietary fats. Given the episodic nature of BA secretion and intestinal re-absorption, the circulating and tissue levels of BAs, like those of the gut hormones, fluctuate in fasting and fed states, and BA levels and forms are markedly affected by disease. BAs exert widespread hormonal-like effects by activating receptors in the nucleus and at the plasma membrane. The nuclear steroid receptors mediate the genomic actions of BAs on BA, glucose and lipid homeostasis. GPBA (TGR5) is a G-protein coupled plasma membrane receptor for BAs that mediates many of the rapid, non-genomic actions of BAs. GPBA has been implicated in the control of glucose homeostasis, inflammation and liver functions. Recent observations have revealed an unexpected role for GPBA in the nervous system. GPBA is expressed by enteric neurons and enterochromaffin cells that control peristalsis, and GPBA mediates the prokinetic actions of BAs in the colon that have been known for millennia. GPBA is also present on primary spinal afferent and spinal neurons that are necessary for sensory transduction. BA-induced activation of GPBA in the sensory nervous system promotes scratching behaviours and analgesia, which may contribute to the pruritus and painless jaundice that are observed in some patients with chronic cholestatic disease, where circulating BA concentrations are markedly increased. Thus, GPBA has emerged as an intriguing target for diverse metabolic, inflammatory, digestive and sensory disorders, where agonists and antagonists may be of value.
Collapse
Affiliation(s)
- T Lieu
- Monash Institute of Pharmaceutical Sciences, Parkville, Vic., Australia
| | | | | |
Collapse
|
1104
|
Aw W, Fukuda S. Toward the comprehensive understanding of the gut ecosystem via metabolomics-based integrated omics approach. Semin Immunopathol 2014; 37:5-16. [PMID: 25338280 DOI: 10.1007/s00281-014-0456-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 10/09/2014] [Indexed: 12/27/2022]
Abstract
Recent advances in DNA sequencing and mass spectrometry technologies have allowed us to collect more data on microbiome and metabolome to assess the influence of the gut microbiota on human health at a whole-systems level. Major advances in metagenomics and metabolomics technologies have shown that the gut microbiota contributes to host overall health status to a large extent. As such, the gut microbiota is often likened to a measurable and functional organ consisting of prokaryotic cells, which creates the unique gut ecosystem together with the host eukaryotic cells. In this review, we discuss in detail the relationship between gut microbiota and its metabolites like choline, bile acids, phenols, and short-chain fatty acids in the host health and etiopathogenesis of various pathological states such as multiple sclerosis, autism, obesity, diabetes, and chronic kidney disease. By integrating metagenomic and metabolomic information on a systems biology-wide approach, we would be better able to understand this interplay between gut microbiome and host metabolism. Integration of the microbiome, metatranscriptome, and metabolome information will pave the way toward an improved holistic understanding of the complex mammalian superorganism. Through the modeling of metabolic interactions between lifestyle, diet, and microbiota, integrated omics-based understanding of the gut ecosystem is the new avenue, providing exciting novel therapeutic approaches for optimal host health.
Collapse
Affiliation(s)
- Wanping Aw
- Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka, Yamagata, 997-0052, Japan
| | | |
Collapse
|
1105
|
Sonne DP, Rehfeld JF, Holst JJ, Vilsbøll T, Knop FK. Postprandial gallbladder emptying in patients with type 2 diabetes: potential implications for bile-induced secretion of glucagon-like peptide 1. Eur J Endocrinol 2014; 171:407-19. [PMID: 24986531 DOI: 10.1530/eje-14-0309] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Recent preclinical work has suggested that postprandial flow of bile acids into the small intestine potentiates nutrient-induced glucagon-like peptide 1 (GLP1(GCG)) secretion via bile acid-induced activation of the G protein-coupled receptor TGR5 in intestinal L cells. The notion of bile-induced GLP1 secretion combined with the findings of reduced postprandial gallbladder emptying in patients with type 2 diabetes (T2DM) led us to speculate whether reduced postprandial GLP1 responses in some patients with T2DM arise as a consequence of diabetic gallbladder dysmotility. DESIGN AND METHODS In a randomised design, 15 patients with long-standing T2DM and 15 healthy age-, gender- and BMI-matched control subjects were studied during 75-g oral glucose tolerance test (OGTT) and three isocaloric (500 kcal) and isovolaemic (350 ml) liquid meals: i) 2.5 g fat, 107 g carbohydrate and 13 g protein; ii) 10 g fat, 93 g carbohydrate and 11 g protein; and iii) 40 g fat, 32 g carbohydrate and 3 g protein. Basal and postprandial plasma concentrations of glucose, insulin, C-peptide, glucagon, GLP1, glucose-dependent insulinotropic polypeptide (GIP), cholecystokinin and gastrin were measured. Furthermore, gallbladder emptying and gastric emptying were examined. RESULTS Gallbladder emptying increased with increasing meal fat content, but no intergroup differences were demonstrated. GIP and GLP1 responses were comparable among the groups with GIP levels being higher following high-fat meals, whereas GLP1 secretion was similar after both OGTT and meals. CONCLUSIONS In conclusion, patients with T2DM exhibited normal gallbladder emptying to meals with a wide range of fat content. Incretin responses were similar to that in controls, and an association with postprandial gallbladder contraction could not be demonstrated.
Collapse
Affiliation(s)
- David P Sonne
- Diabetes Research DivisionDepartment of Medicine, Gentofte Hospital, University of Copenhagen, Niels Andersens Vej 65, DK-2900 Hellerup, DenmarkThe NNF Center for Basic Metabolic ResearchDepartment of Biomedical Sciences, The Panum Institute, University of Copenhagen, Copenhagen, DenmarkDepartment of Clinical BiochemistryCopenhagen University Hospital Rigshospitalet, Copenhagen, Denmark Diabetes Research DivisionDepartment of Medicine, Gentofte Hospital, University of Copenhagen, Niels Andersens Vej 65, DK-2900 Hellerup, DenmarkThe NNF Center for Basic Metabolic ResearchDepartment of Biomedical Sciences, The Panum Institute, University of Copenhagen, Copenhagen, DenmarkDepartment of Clinical BiochemistryCopenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Jens F Rehfeld
- Diabetes Research DivisionDepartment of Medicine, Gentofte Hospital, University of Copenhagen, Niels Andersens Vej 65, DK-2900 Hellerup, DenmarkThe NNF Center for Basic Metabolic ResearchDepartment of Biomedical Sciences, The Panum Institute, University of Copenhagen, Copenhagen, DenmarkDepartment of Clinical BiochemistryCopenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Jens J Holst
- Diabetes Research DivisionDepartment of Medicine, Gentofte Hospital, University of Copenhagen, Niels Andersens Vej 65, DK-2900 Hellerup, DenmarkThe NNF Center for Basic Metabolic ResearchDepartment of Biomedical Sciences, The Panum Institute, University of Copenhagen, Copenhagen, DenmarkDepartment of Clinical BiochemistryCopenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Tina Vilsbøll
- Diabetes Research DivisionDepartment of Medicine, Gentofte Hospital, University of Copenhagen, Niels Andersens Vej 65, DK-2900 Hellerup, DenmarkThe NNF Center for Basic Metabolic ResearchDepartment of Biomedical Sciences, The Panum Institute, University of Copenhagen, Copenhagen, DenmarkDepartment of Clinical BiochemistryCopenhagen University Hospital Rigshospitalet, Copenhagen, Denmark Diabetes Research DivisionDepartment of Medicine, Gentofte Hospital, University of Copenhagen, Niels Andersens Vej 65, DK-2900 Hellerup, DenmarkThe NNF Center for Basic Metabolic ResearchDepartment of Biomedical Sciences, The Panum Institute, University of Copenhagen, Copenhagen, DenmarkDepartment of Clinical BiochemistryCopenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Filip K Knop
- Diabetes Research DivisionDepartment of Medicine, Gentofte Hospital, University of Copenhagen, Niels Andersens Vej 65, DK-2900 Hellerup, DenmarkThe NNF Center for Basic Metabolic ResearchDepartment of Biomedical Sciences, The Panum Institute, University of Copenhagen, Copenhagen, DenmarkDepartment of Clinical BiochemistryCopenhagen University Hospital Rigshospitalet, Copenhagen, Denmark Diabetes Research DivisionDepartment of Medicine, Gentofte Hospital, University of Copenhagen, Niels Andersens Vej 65, DK-2900 Hellerup, DenmarkThe NNF Center for Basic Metabolic ResearchDepartment of Biomedical Sciences, The Panum Institute, University of Copenhagen, Copenhagen, DenmarkDepartment of Clinical BiochemistryCopenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
1106
|
Abstract
Bile acids are the end products of cholesterol catabolism. Hepatic bile acid synthesis accounts for a major fraction of daily cholesterol turnover in humans. Biliary secretion of bile acids generates bile flow and facilitates hepatobiliary secretion of lipids, lipophilic metabolites, and xenobiotics. In the intestine, bile acids are essential for the absorption, transport, and metabolism of dietary fats and lipid-soluble vitamins. Extensive research in the last 2 decades has unveiled new functions of bile acids as signaling molecules and metabolic integrators. The bile acid-activated nuclear receptors farnesoid X receptor, pregnane X receptor, constitutive androstane receptor, vitamin D receptor, and G protein-coupled bile acid receptor play critical roles in the regulation of lipid, glucose, and energy metabolism, inflammation, and drug metabolism and detoxification. Bile acid synthesis exhibits a strong diurnal rhythm, which is entrained by fasting and refeeding as well as nutrient status and plays an important role for maintaining metabolic homeostasis. Recent research revealed an interaction of liver bile acids and gut microbiota in the regulation of liver metabolism. Circadian disturbance and altered gut microbiota contribute to the pathogenesis of liver diseases, inflammatory bowel diseases, nonalcoholic fatty liver disease, diabetes, and obesity. Bile acids and their derivatives are potential therapeutic agents for treating metabolic diseases of the liver.
Collapse
Affiliation(s)
- Tiangang Li
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (T.L.); and Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio (J.Y.L.C.)
| | - John Y L Chiang
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas (T.L.); and Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio (J.Y.L.C.)
| |
Collapse
|
1107
|
Abstract
Obesity and related diseases are a major cause of human morbidity and mortality and constitute a substantial economic burden for society. Effective treatment regimens are scarce, and new therapeutic targets are needed. Brown adipose tissue, an energy-expending tissue that produces heat, represents a potential therapeutic target. Its presence is associated with low body mass index, low total adipose tissue content and a lower risk of type 2 diabetes mellitus. Knowledge about the development and function of thermogenic adipocytes in brown adipose tissue has increased substantially in the last decade. Important transcriptional regulators have been identified, and hormones able to modulate the thermogenic capacity of the tissue have been recognized. Intriguingly, it is now clear that humans, like rodents, possess two types of thermogenic adipocytes: the classical brown adipocytes found in the interscapular brown adipose organ and the so-called beige adipocytes primarily found in subcutaneous white adipose tissue after adrenergic stimulation. The presence of two distinct types of energy-expending adipocytes in humans is conceptually important because these cells might be stimulated and recruited by different signals, raising the possibility that they might be separate potential targets for therapeutic intervention. In this review, we will discuss important features of the energy-expending brown adipose tissue and highlight those that may serve as potential targets for pharmacological intervention aimed at expanding the tissue and/or enhancing its function to counteract obesity.
Collapse
Affiliation(s)
- M E Lidell
- Department of Medical and Clinical Genetics, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | | |
Collapse
|
1108
|
Gioiello A, Cerra B, Zhang W, Vallerini GP, Costantino G, De Franco F, Passeri D, Pellicciari R, Setchell KDR. Synthesis of atypical bile acids for use as investigative tools for the genetic defect of 3β-hydroxy-Δ(5)-C27-steroid oxidoreductase deficiency. J Steroid Biochem Mol Biol 2014; 144 Pt B:348-60. [PMID: 24954360 DOI: 10.1016/j.jsbmb.2014.06.008] [Citation(s) in RCA: 9] [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: 03/27/2014] [Revised: 05/28/2014] [Accepted: 06/17/2014] [Indexed: 12/31/2022]
Abstract
Deficiency of 3β-hydroxy-Δ(5)-C27-steroid oxidoreductase (HSD3B7), an enzyme catalyzing the second step in the pathway for bile acid synthesis, leads to a complete lack of the primary bile acids, cholic and chenodeoxycholic acids, and the accumulation of 3β,7α-dihydroxy- and 3β,7α,12α-trihydroxy-Δ(5)-cholenoic acids. Patients affected by this autosomal recessive genetic defect develop cholestatic liver disease that is clinically responsive to primary bile acid therapy. Reference standards of these compounds are needed to facilitate diagnosis and to accurately quantify biochemical responses to therapy. Described are a novel synthesis of atypical bile acids that characterize the HSD3B7 deficiency and their effect on bile acid-activated nuclear receptors, target genes and cytochromes involved in bile acid homeostasis and detoxification. The failure of 3β-hydroxy-Δ(5)-cholenoic acids to function as FXR, PXR and CAR agonists and to exert hepatoprotective actions explains the mechanism for progressive cholestatic liver disease in patients with HSD3B7 deficiency.
Collapse
Affiliation(s)
- Antimo Gioiello
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo 1, I-06122 Perugia, Italy.
| | - Bruno Cerra
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo 1, I-06122 Perugia, Italy
| | - Wujuan Zhang
- Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center and Department of Pediatrics of the University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Gian Paolo Vallerini
- Department of Farmacy, University of Parma, Viale delle Scienze 27/A, Parma I-43124, Italy
| | - Gabriele Costantino
- Department of Farmacy, University of Parma, Viale delle Scienze 27/A, Parma I-43124, Italy
| | | | - Daniela Passeri
- TES Pharma, Via P. Togliatti, 20, Loc Taverne, I-06073 Corciano, Italy
| | - Roberto Pellicciari
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo 1, I-06122 Perugia, Italy; TES Pharma, Via P. Togliatti, 20, Loc Taverne, I-06073 Corciano, Italy
| | - Kenneth D R Setchell
- Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center and Department of Pediatrics of the University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
1109
|
Domingue JC, Ao M, Sarathy J, George A, Alrefai WA, Nelson DJ, Rao MC. HEK-293 cells expressing the cystic fibrosis transmembrane conductance regulator (CFTR): a model for studying regulation of Cl- transport. Physiol Rep 2014; 2:2/9/e12158. [PMID: 25263207 PMCID: PMC4270233 DOI: 10.14814/phy2.12158] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The Human Embryonic Kidney 293 cell line (HEK‐293) readily lends itself to genetic manipulation and is a common tool for biologists to overexpress proteins of interest and study their function and molecular regulation. Although these cells have some limitations, such as an inability to form resistive monolayers necessary for studying transepithelial ion transport, they are nevertheless valuable in studying individual epithelial ion transporters. We report the use of HEK‐293 cells to study the cystic fibrosis transmembrane conductance regulator (CFTR) Cl− channel. While HEK‐293 cells endogenously express mRNA for the Cl− channels, ClC‐2 and TMEM16A, they neither express CFTR mRNA nor protein. Therefore, we stably transfected HEK‐293 cells with EGFP‐CFTR (HEK‐CFTR) and demonstrated CFTR function by measuring forskolin‐stimulated iodide efflux. This efflux was inhibited by CFTRinh172, and the protein kinase A inhibitor H89, but not by Ca2+ chelation. In contrast to intestinal epithelia, forskolin stimulation does not increase surface CFTR expression and does not require intact microtubules in HEK‐CFTR. To investigate the role of an endogenous GαS‐coupled receptor, we examined the bile acid receptor, TGR5. Although HEK‐CFTR cells express TGR5, the potent TGR5 agonist lithocholic acid (LCA; 5–500 μmol/L) did not activate CFTR. Furthermore, forskolin, but not LCA, increased [cAMP]i in HEK‐CFTR suggesting that endogenous TGR5 may not be functionally linked to GαS. However, LCA did increase [Ca2+]i and interestingly, abolished forskolin‐stimulated iodide efflux. Thus, we propose that the stable HEK‐CFTR cell line is a useful model to study the multiple signaling pathways that regulate CFTR. In this study, we characterize a HEK‐293 cell line, stably transfected with EGFP‐CFTR (HEK‐CFTR). We examined its regulation by endogenously expressed signaling pathways, in particular the cAMP and the GαS‐coupled bile acid receptor, TGR5, signaling pathways.
Collapse
Affiliation(s)
- Jada C Domingue
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois
| | - Mei Ao
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois
| | - Jayashree Sarathy
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois Department of Biological Sciences, Benedictine University, Lisle, Illinois
| | - Alvin George
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois
| | - Waddah A Alrefai
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois Jesse Brown V.A. Medical Center, Chicago, Illinois
| | - Deborah J Nelson
- Department of Pharmacological and Physiological Sciences, The University of Chicago, Chicago, Illinois
| | - Mrinalini C Rao
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
1110
|
De Giorgi S, Campos V, Egli L, Toepel U, Carrel G, Cariou B, Rainteau D, Schneiter P, Tappy L, Giusti V. Long-term effects of Roux-en-Y gastric bypass on postprandial plasma lipid and bile acids kinetics in female non diabetic subjects: A cross-sectional pilot study. Clin Nutr 2014; 34:911-7. [PMID: 25306425 DOI: 10.1016/j.clnu.2014.09.018] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 08/25/2014] [Accepted: 09/22/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Formerly obese patients having undergone Roux-en-Y gastric bypass (RYGB) display both an accelerated digestion and absorption of carbohydrate and an increased plasma glucose clearance rate after meal ingestion. How RYGB effects postprandial kinetics of dietary lipids has yet not been investigated. METHODS Plasma triglyceride (TG), apoB48, total apoB, bile acids (BA), fibroblast growth factor 19 (FGF19), and cholecystokinin (CCK) were measured in post-absorptive conditions and over 4-h following the ingestion of a mixed test meal in a cross-sectional, pilot study involving 11 formerly obese female patients 33.8 ± 16.4 months after RYGB surgery and in 11 weight- and age-matched female control participants. RESULTS Compared to controls, RYGB patients had faster (254 ± 14 vs. 327 ± 7 min, p < 0.05) and lower (0.14 ± 0.04 vs. 0.35 ± 0.07 mM, p < 0.05) peak TG responses, but their peak apoB48 responses tended to be higher (2692 ± 336 vs. 1841 ± 228 ng/ml, p = 0.09). Their postprandial total BA concentrations were significantly increased and peaked earlier after meal ingestion than in controls. Their FGF19 and CCK concentrations also peaked earlier and to a higher value. CONCLUSIONS The early postprandial apoB48 and BA responses indicate that RYGB accelerated the rate of dietary lipid absorption. The lower postprandial peak TG strongly suggests that the RYGB simultaneously increased the clearance of TG-rich lipoproteins. CLINICAL TRIAL REGISTRATION NCT01891591.
Collapse
Affiliation(s)
- Sara De Giorgi
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne, Switzerland
| | - Vanessa Campos
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne, Switzerland
| | - Leonie Egli
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne, Switzerland
| | - Ulrike Toepel
- Departments of Radiology and Clinical Neurosciences, University Hospital and University of Lausanne, Switzerland
| | - Guillaume Carrel
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne, Switzerland
| | - Bertrand Cariou
- INSERM, UMR1087-CNRS UMR6291, l'Institut du Thorax, Faculté de Médecine, Université de Nantes, F-44000 Nantes, France
| | - Dominique Rainteau
- Sorbonne Universités - UPMC Univ Paris 06, INSERM ERL 1157, CNRS UMR 7203 LBM, CHU Saint-Antoine, Paris, France
| | - Philippe Schneiter
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne, Switzerland
| | - Luc Tappy
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne, Switzerland.
| | - Vittorio Giusti
- Center for Metabolic Diseases, Department of Internal Medicine, Broye Intercantonal Hospital, Estavayer-le-lac, Switzerland
| |
Collapse
|
1111
|
Lutz TA, Bueter M. The physiology underlying Roux-en-Y gastric bypass: a status report. Am J Physiol Regul Integr Comp Physiol 2014; 307:R1275-91. [PMID: 25253084 DOI: 10.1152/ajpregu.00185.2014] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Obesity and its related comorbidities can be detrimental for the affected individual and challenge public health systems worldwide. Currently, the only available treatment options leading to clinically significant and maintained body weight loss and reduction in obesity-related morbidity and mortality are based on surgical interventions. This review will focus on two main clinical effects of Roux-en-Y gastric bypass (RYGB), namely body weight loss and change in eating behavior. Animal experiments designed to understand the underlying physiological mechanisms of these post-gastric bypass effects will be discussed. Where appropriate, reference will also be made to vertical sleeve gastrectomy. While caloric malabsorption and mechanical restriction seem not to be major factors in this respect, alterations in gut hormone levels are invariably found after RYGB. However, their causal role in RYGB effects on eating and body weight has recently been challenged. Other potential factors contributing to the RYGB effects include increased bile acid concentrations and an altered composition of gut microbiota. RYGB is further associated with remarkable changes in preference for different dietary components, such as a decrease in the preference for high fat or sugar. It needs to be noted, however, that in many cases, the question about the necessity of these alterations for the success of bariatric surgery procedures remains unanswered.
Collapse
Affiliation(s)
- Thomas A Lutz
- Institute of Veterinary Physiology, Vetsuisse Faculty University of Zurich, Zurich, Switzerland; Center of Integrative Human Physiology, University of Zurich, Zurich, Switzerland; Institute of Laboratory Animal Science, University of Zurich, Zurich, Switzerland; and
| | - Marco Bueter
- Center of Integrative Human Physiology, University of Zurich, Zurich, Switzerland; Department of Surgery, Division of Visceral and Transplantation Surgery, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
1112
|
Dual activation of the bile acid nuclear receptor FXR and G-protein-coupled receptor TGR5 protects mice against atherosclerosis. PLoS One 2014; 9:e108270. [PMID: 25237811 PMCID: PMC4169583 DOI: 10.1371/journal.pone.0108270] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 08/26/2014] [Indexed: 12/17/2022] Open
Abstract
Bile acid signaling is a critical regulator of glucose and energy metabolism, mainly through the nuclear receptor FXR and the G protein-coupled receptor TGR. The purpose of the present study was to investigate whether dual activation of FXR and TGR5 plays a significant role in the prevention of atherosclerosis progression. To evaluate the effects of bile acid signaling in atherogenesis, ApoE-/- mice and LDLR-/- mice were treated with an FXR/TGR5 dual agonist (INT-767). INT-767 treatment drastically reduced serum cholesterol levels. INT-767 treatment significantly reduced atherosclerotic plaque formation in both ApoE-/- and LDLR-/- mice. INT-767 decreased the expression of pro-inflammatory cytokines and chemokines in the aortas of ApoE-/- mice through the inactivation of NF-κB. In addition, J774 macrophages treated with INT-767 had significantly lower levels of active NF-κB, resulting in cytokine production in response to LPS through a PKA dependent mechanism. This study demonstrates that concurrent activation of FXR and TGR5 attenuates atherosclerosis by reducing both circulating lipids and inflammation.
Collapse
|
1113
|
Ullmer C, Alvarez Sanchez R, Sprecher U, Raab S, Mattei P, Dehmlow H, Sewing S, Iglesias A, Beauchamp J, Conde-Knape K. Systemic bile acid sensing by G protein-coupled bile acid receptor 1 (GPBAR1) promotes PYY and GLP-1 release. Br J Pharmacol 2014; 169:671-84. [PMID: 23488746 DOI: 10.1111/bph.12158] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 01/18/2013] [Accepted: 02/07/2013] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE Nutrient sensing in the gut is believed to be accomplished through activation of GPCRs expressed on enteroendocrine cells. In particular, L-cells located predominantly in distal regions of the gut secrete glucagon-like peptide 1 (GLP-1) and peptide tyrosine-tyrosine (PYY) upon stimulation by nutrients and bile acids (BA). The study was designed to address the mechanism of hormone secretion in L-cells stimulated by the BA receptor G protein-coupled bile acid receptor 1 (GPBAR1). EXPERIMENTAL APPROACH A novel, selective, orally bioavailable, and potent GPBAR1 agonist, RO5527239, was synthesized in order to investigate L-cell secretion in vitro and in vivo in mice and monkey. In analogy to BA, RO5527239 was conjugated with taurine to reduce p.o. bioavailability yet retaining its potency. Using RO5527239 and tauro-RO5527239, the acute secretion effects on L-cells were addressed via different routes of administration. KEY RESULTS GPBAR1 signalling triggers the co-secretion of PYY and GLP-1, and leads to improved glucose tolerance. The strong correlation of plasma drug exposure and plasma PYY levels suggests activation of GPBAR1 from systemically accessible compartments. In contrast to the orally bioavailable agonist RO5527239, we show that tauro-RO5527239 triggers PYY release only when applied intravenously. Compared to mice, a slower and more sustained PYY secretion was observed in monkeys. CONCLUSION AND IMPLICATIONS Selective GPBAR1 activation elicits a strong secretagogue effect on L-cells, which primarily requires systemic exposure. We suggest that GPBAR1 is a key player in the intestinal proximal-distal loop that mediates the early phase of nutrient-evoked L-cell secretion effects.
Collapse
Affiliation(s)
- C Ullmer
- DTA CV & Metabolic Diseases, DTA CV and Metabolic Diseases, Pharma Research and Early Development, F. Hoffmann-La Roche AG, Basel, Switzerland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1114
|
Sepe V, Renga B, Festa C, D'Amore C, Masullo D, Cipriani S, Di Leva FS, Monti MC, Novellino E, Limongelli V, Zampella A, Fiorucci S. Modification on ursodeoxycholic acid (UDCA) scaffold. discovery of bile acid derivatives as selective agonists of cell-surface G-protein coupled bile acid receptor 1 (GP-BAR1). J Med Chem 2014; 57:7687-701. [PMID: 25162837 DOI: 10.1021/jm500889f] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Bile acids are signaling molecules interacting with the nuclear receptor FXR and the G-protein coupled receptor 1 (GP-BAR1/TGR5). GP-BAR1 is a promising pharmacological target for the treatment of steatohepatitis, type 2 diabetes, and obesity. Endogenous bile acids and currently available semisynthetic bile acids are poorly selective toward GP-BAR1 and FXR. Thus, in the present study we have investigated around the structure of UDCA, a clinically used bile acid devoid of FXR agonist activity, to develop a large family of side chain modified 3α,7β-dihydroxyl cholanoids that selectively activate GP-BAR1. In vivo and in vitro pharmacological evaluation demonstrated that administration of compound 16 selectively increases the expression of pro-glucagon 1, a GP-BAR1 target, in the small intestine, while it had no effect on FXR target genes in the liver. Further, compound 16 results in a significant reshaping of bile acid pool in a rodent model of cholestasis. These data demonstrate that UDCA is a useful scaffold to generate novel and selective steroidal ligands for GP-BAR1.
Collapse
Affiliation(s)
- Valentina Sepe
- Department of Pharmacy, University of Naples "Federico II" , Via D. Montesano, 49, 80131 Napoli, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1115
|
Discovery of novel pyrimidine and malonamide derivatives as TGR5 agonists. Bioorg Med Chem Lett 2014; 24:4271-5. [DOI: 10.1016/j.bmcl.2014.07.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 06/17/2014] [Accepted: 07/09/2014] [Indexed: 12/31/2022]
|
1116
|
Rajan S, Gupta A, Beg M, Shankar K, Srivastava A, Varshney S, Kumar D, Gaikwad AN. Adipocyte transdifferentiation and its molecular targets. Differentiation 2014; 87:183-92. [PMID: 25130315 DOI: 10.1016/j.diff.2014.07.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 07/02/2014] [Accepted: 07/24/2014] [Indexed: 01/19/2023]
Abstract
According to the World Health Organization obesity is defined as the excessive accumulation of fat, which increases risk of other metabolic disorders such as insulin resistance, dyslipidemia, hypertension, cardiovascular diseases, etc. There are two types of adipose tissue, white and brown adipose tissue (BAT) and the latter has recently gathered interest of the scientific community. Discovery of BAT has opened avenues for a new therapeutic strategy for the treatment of obesity and related metabolic syndrome. BAT utilizes accumulated fatty acids for energy expenditure; hence it is seen as one of the possible alternates to the current treatment. Moreover, browning of white adipocyte on exposure to cold, as well as with some of the pharmacological agents presents exciting outcomes and indicates the feasibility of transdifferentiation. A better understanding of molecular pathways and differentiation factors, those that play a key role in transdifferentiation are of extreme importance in designing novel strategies for the treatment of obesity and associated metabolic disorders.
Collapse
Affiliation(s)
- Sujith Rajan
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031 UP, India; Academy of Scientific and Innovative Research, CSIR-CDRI, India
| | - Abhishek Gupta
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031 UP, India
| | - Muheeb Beg
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031 UP, India
| | - Kripa Shankar
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031 UP, India
| | - Ankita Srivastava
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031 UP, India; Academy of Scientific and Innovative Research, CSIR-CDRI, India
| | - Salil Varshney
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031 UP, India
| | - Durgesh Kumar
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031 UP, India; Academy of Scientific and Innovative Research, CSIR-CDRI, India
| | - Anil Nilkanth Gaikwad
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031 UP, India; Academy of Scientific and Innovative Research, CSIR-CDRI, India.
| |
Collapse
|
1117
|
Udayappan SD, Hartstra AV, Dallinga-Thie GM, Nieuwdorp M. Intestinal microbiota and faecal transplantation as treatment modality for insulin resistance and type 2 diabetes mellitus. Clin Exp Immunol 2014; 177:24-9. [PMID: 24528224 DOI: 10.1111/cei.12293] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2014] [Indexed: 12/17/2022] Open
Abstract
The prevalence of obesity and diabetes mellitus type 2 is increasing rapidly around the globe. Recent insights have generated an entirely new perspective that the intestinal microbiota may play a significant role in the development of these metabolic disorders. Alterations in the intestinal microbiota composition promote systemic inflammation that is a hallmark of obesity and subsequent insulin resistance. Thus, it is important to understand the reciprocal relationship between intestinal microbiota composition and metabolic health in order to eventually prevent disease progression. In this respect, faecal transplantation studies have implicated that butyrate-producing intestinal bacteria are crucial in this process and be considered as key players in regulating diverse signalling cascades associated with human glucose and lipid metabolism.
Collapse
Affiliation(s)
- S D Udayappan
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | | | | | | |
Collapse
|
1118
|
Sonne DP, Hansen M, Knop FK. Bile acid sequestrants in type 2 diabetes: potential effects on GLP1 secretion. Eur J Endocrinol 2014; 171:R47-65. [PMID: 24760535 DOI: 10.1530/eje-14-0154] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Bile acid sequestrants have been used for decades for the treatment of hypercholesterolaemia. Sequestering of bile acids in the intestinal lumen interrupts enterohepatic recirculation of bile acids, which initiate feedback mechanisms on the conversion of cholesterol into bile acids in the liver, thereby lowering cholesterol concentrations in the circulation. In the early 1990s, it was observed that bile acid sequestrants improved glycaemic control in patients with type 2 diabetes. Subsequently, several studies confirmed the finding and recently - despite elusive mechanisms of action - bile acid sequestrants have been approved in the USA for the treatment of type 2 diabetes. Nowadays, bile acids are no longer labelled as simple detergents necessary for lipid digestion and absorption, but are increasingly recognised as metabolic regulators. They are potent hormones, work as signalling molecules on nuclear receptors and G protein-coupled receptors and trigger a myriad of signalling pathways in many target organs. The most described and well-known receptors activated by bile acids are the farnesoid X receptor (nuclear receptor) and the G protein-coupled cell membrane receptor TGR5. Besides controlling bile acid metabolism, these receptors are implicated in lipid, glucose and energy metabolism. Interestingly, activation of TGR5 on enteroendocrine L cells has been suggested to affect secretion of incretin hormones, particularly glucagon-like peptide 1 (GLP1 (GCG)). This review discusses the role of bile acid sequestrants in the treatment of type 2 diabetes, the possible mechanism of action and the role of bile acid-induced secretion of GLP1 via activation of TGR5.
Collapse
Affiliation(s)
- David P Sonne
- Diabetes Research DivisionDepartment of Medicine, Gentofte Hospital, Niels Andersens Vej 65, DK-2900 Hellerup, Denmark
| | - Morten Hansen
- Diabetes Research DivisionDepartment of Medicine, Gentofte Hospital, Niels Andersens Vej 65, DK-2900 Hellerup, Denmark
| | - Filip K Knop
- Diabetes Research DivisionDepartment of Medicine, Gentofte Hospital, Niels Andersens Vej 65, DK-2900 Hellerup, Denmark
| |
Collapse
|
1119
|
Penno CA, Morgan SA, Rose AJ, Herzig S, Lavery GG, Odermatt A. 11β-Hydroxysteroid dehydrogenase-1 is involved in bile acid homeostasis by modulating fatty acid transport protein-5 in the liver of mice. Mol Metab 2014; 3:554-64. [PMID: 25061560 PMCID: PMC4099504 DOI: 10.1016/j.molmet.2014.04.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 04/17/2014] [Accepted: 04/19/2014] [Indexed: 12/31/2022] Open
Abstract
11β-Hydroxysteroid dehydrogenase-1 (11β-HSD1) plays a key role in glucocorticoid receptor (GR) activation. Besides, it metabolizes some oxysterols and bile acids (BAs). The GR regulates BA homeostasis; however, the impact of impaired 11β-HSD1 activity remained unknown. We profiled plasma and liver BAs in liver-specific and global 11β-HSD1-deficient mice. 11β-HSD1-deficiency resulted in elevated circulating unconjugated BAs, an effect more pronounced in global than liver-specific knockout mice. Gene expression analyses revealed decreased expression of the BA-CoA ligase Fatp5, suggesting impaired BA amidation. Reduced organic anion-transporting polypeptide-1A1 (Oatp1a1) and enhanced organic solute-transporter-β (Ostb) mRNA expression were observed in livers from global 11β-HSD1-deficient mice. The impact of 11β-HSD1-deficiency on BA homeostasis seems to be GR-independent because intrahepatic corticosterone and GR target gene expression were not substantially decreased in livers from global knockout mice. Moreover, Fatp5 expression in livers from hepatocyte-specific GR knockout mice was unchanged. The results revealed a role for 11β-HSD1 in BA homeostasis.
Collapse
Key Words
- 11β-Hydroxysteroid dehydrogenase
- 11β-hydroxysteroid dehydrogenase 1, 11β-HSD1
- BA coenzyme A: amino acid N-acyltransferase, Baat
- Bile acid conjugation
- Bile acid transport
- Bile acids
- Glucocorticoids
- Na+-taurocholate cotransporting polypeptide, Ntcp
- Organic anion-transporting polypeptide, Oatp
- Organic solute transporter, Ost
- bile acids, BAs
- cholesterol 7α-hydroxylase, Cyp7a1
- farnesoid X receptor, Fxr
- fatty acid transport protein, Fatp
- glucocorticoid receptor, GR
- short heterodimer partner, Shp
- sterol-regulatory element-binding protein 1C, Srebp1c
Collapse
Affiliation(s)
- Carlos A. Penno
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Stuart A. Morgan
- Centre for Endocrinology Diabetes and Metabolism (CEDAM), Institute of Biomedical Research, Medical School Building, School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Adam J. Rose
- Joint Research Division, Molecular Metabolic Control, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH), Heidelberg University, Network Aging Research, University Hospital Heidelberg, Germany
| | - Stephan Herzig
- Joint Research Division, Molecular Metabolic Control, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH), Heidelberg University, Network Aging Research, University Hospital Heidelberg, Germany
| | - Gareth G. Lavery
- Centre for Endocrinology Diabetes and Metabolism (CEDAM), Institute of Biomedical Research, Medical School Building, School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Alex Odermatt
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| |
Collapse
|
1120
|
Abstract
Over the past decade, it has become apparent that bile acids are involved in a host of activities beyond their classic functions in bile formation and fat absorption. The identification of the farnesoid X receptor (FXR) as a nuclear receptor directly activated by bile acids and the discovery that bile acids are also ligands for the membrane-bound, G-protein coupled bile acid receptor 1 (also known as TGR5) have opened new avenues of research. Both FXR and TGR5 regulate various elements of glucose, lipid and energy metabolism. Consequently, a picture has emerged of bile acids acting as modulators of (postprandial) metabolism. Therefore, strategies that interfere with either bile acid metabolism or signalling cascades mediated by bile acids may represent novel therapeutic approaches for metabolic diseases. Synthetic modulators of FXR have been designed and tested, primarily in animal models. Furthermore, the use of bile acid sequestrants to reduce plasma cholesterol levels has unexpected benefits. For example, treatment of patients with type 2 diabetes mellitus (T2DM) with sequestrants causes substantial reductions in plasma levels of glucose and HbA1c. This Review aims to provide an overview of the molecular mechanisms by which bile acids modulate glucose and energy metabolism, particularly focusing on the glucose-lowering actions of bile acid sequestrants in insulin resistant states and T2DM.
Collapse
Affiliation(s)
- Folkert Kuipers
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700RB Groningen, Netherlands
| | - Vincent W Bloks
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700RB Groningen, Netherlands
| | - Albert K Groen
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700RB Groningen, Netherlands
| |
Collapse
|
1121
|
Appleby RN, Walters JRF. The role of bile acids in functional GI disorders. Neurogastroenterol Motil 2014; 26:1057-69. [PMID: 24898156 DOI: 10.1111/nmo.12370] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 04/28/2014] [Indexed: 02/06/2023]
Abstract
BACKGROUND Bile acids are increasingly implicated in the pathogenesis of functional GI disorders. New mechanisms have recently been described in the irritable bowel syndrome, chronic diarrhea and chronic idiopathic constipation. Identification of bile acid signaling through farnesoid X receptor (FXR), transmembrane G-coupled receptor 5 (TGR5) and fibroblast growth factor 19 (FGF19) has led to the development of new, directly acting therapeutic agents. Despite these advances primary bile acid diarrhea remains under-recognized partly because of the lack of a widely available diagnostic test. PURPOSE In this review we will summarize the effects of bile acids on bowel function throughout the gastrointestinal tract and their roles in the pathogenesis of functional diseases. We will review established diagnostic tests and therapies for functional heartburn, dyspepsia and bile acid diarrhea. There will be a particular emphasis on recent trial data for emerging therapies such as Elobixibat and Obeticholic acid and novel diagnostic tests for bile acid diarrhea such as 7α-Hydroxy-4-cholesten-3-one (C4) and FGF19. Finally we will discuss future directions for research in this rapidly evolving field, such as bacterial bile acid modification and identification of genetic anomalies associated with functional disorders.
Collapse
Affiliation(s)
- Richard N Appleby
- Section of Hepatology and Gastroenterology, Imperial College London, Imperial College Healthcare, Hammersmith Hospital, London, UK
| | | |
Collapse
|
1122
|
Sweeney TE, Morton JM. Metabolic surgery: action via hormonal milieu changes, changes in bile acids or gut microbiota? A summary of the literature. Best Pract Res Clin Gastroenterol 2014; 28:727-40. [PMID: 25194186 PMCID: PMC4399638 DOI: 10.1016/j.bpg.2014.07.016] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 07/21/2014] [Accepted: 07/28/2014] [Indexed: 01/31/2023]
Abstract
Obesity and type 2 diabetes remain epidemic problems. Different bariatric surgical techniques causes weight loss and diabetes remission to varying degrees. The underlying mechanisms of the beneficial effects of bariatric surgery are complex, and include changes in diet and behaviour, as well as changes in hormones, bile acid flow, and gut bacteria. We summarized the effects of multiple different bariatric procedures, and their resulting effects on several hormones (leptin, ghrelin, adiponectin, glucagon-like peptide 1 (GLP-1), peptide YY, and glucagon), bile acid changes in the gut and the serum, and resulting changes to the gut microbiome. As much as possible, we have tried to incorporate multiple studies to try to explain underlying mechanistic changes. What emerges from the data is a picture of clear differences between restrictive and metabolic procedures. The latter, in particular the roux-en-Y gastric bypass, induces large and distinctive changes in most measured fat and gut hormones, including early and sustained increase in GLP-1, possible through intestinal bile acid signalling. The changes in bile flow and the gut microbiome are causally inseparable so far, but new studies show that each contributes to the effects of weight loss and diabetes resolution.
Collapse
Affiliation(s)
- Timothy E Sweeney
- Stanford University, Department of General Surgery, Section of Bariatric and Minimally Invasive (BMI) Surgery, 300 Pasteur Drive, H3680, Stanford, CA 94025, USA
| | - John M Morton
- Stanford University, Department of General Surgery, Section of Bariatric and Minimally Invasive (BMI) Surgery, 300 Pasteur Drive, H3680, Stanford, CA 94025, USA.
| |
Collapse
|
1123
|
Napolitano A, Miller S, Nicholls AW, Baker D, Van Horn S, Thomas E, Rajpal D, Spivak A, Brown JR, Nunez DJ. Novel gut-based pharmacology of metformin in patients with type 2 diabetes mellitus. PLoS One 2014; 9:e100778. [PMID: 24988476 PMCID: PMC4079657 DOI: 10.1371/journal.pone.0100778] [Citation(s) in RCA: 222] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Accepted: 05/23/2014] [Indexed: 12/20/2022] Open
Abstract
UNLABELLED Metformin, a biguanide derivate, has pleiotropic effects beyond glucose reduction, including improvement of lipid profiles and lowering microvascular and macrovascular complications associated with type 2 diabetes mellitus (T2DM). These effects have been ascribed to adenosine monophosphate-activated protein kinase (AMPK) activation in the liver and skeletal muscle. However, metformin effects are not attenuated when AMPK is knocked out and intravenous metformin is less effective than oral medication, raising the possibility of important gut pharmacology. We hypothesized that the pharmacology of metformin includes alteration of bile acid recirculation and gut microbiota resulting in enhanced enteroendocrine hormone secretion. In this study we evaluated T2DM subjects on and off metformin monotherapy to characterize the gut-based mechanisms of metformin. Subjects were studied at 4 time points: (i) at baseline on metformin, (ii) 7 days after stopping metformin, (iii) when fasting blood glucose (FBG) had risen by 25% after stopping metformin, and (iv) when FBG returned to baseline levels after restarting the metformin. At these timepoints we profiled glucose, insulin, gut hormones (glucagon-like peptide-1 (GLP-1), peptide tyrosine-tyrosine (PYY) and glucose-dependent insulinotropic peptide (GIP) and bile acids in blood, as well as duodenal and faecal bile acids and gut microbiota. We found that metformin withdrawal was associated with a reduction of active and total GLP-1 and elevation of serum bile acids, especially cholic acid and its conjugates. These effects reversed when metformin was restarted. Effects on circulating PYY were more modest, while GIP changes were negligible. Microbiota abundance of the phylum Firmicutes was positively correlated with changes in cholic acid and conjugates, while Bacteroidetes abundance was negatively correlated. Firmicutes and Bacteroidetes representation were also correlated with levels of serum PYY. Our study suggests that metformin has complex effects due to gut-based pharmacology which might provide insights into novel therapeutic approaches to treat T2DM and associated metabolic diseases. TRIAL REGISTRATION www.ClinicalTrials.gov NCT01357876.
Collapse
Affiliation(s)
| | - Sam Miller
- Quantitative Sciences, GSK R&D, Stevenage, Herts, United Kingdom
| | | | - David Baker
- Safety Assessment, GSK R&D, Ware, Herts, United Kingdom
| | - Stephanie Van Horn
- Target and Pathways Validation, GSK R&D, Upper Providence, Pennsylvania, United States of America
| | - Elizabeth Thomas
- Target and Pathways Validation, GSK R&D, Upper Providence, Pennsylvania, United States of America
| | - Deepak Rajpal
- Computational Biology, GSK R&D, Upper Providence, Pennsylvania, United States of America
| | - Aaron Spivak
- Computational Biology, GSK R&D, Upper Providence, Pennsylvania, United States of America
| | - James R. Brown
- Computational Biology, GSK R&D, Upper Providence, Pennsylvania, United States of America
| | - Derek J. Nunez
- Enteroendocrine Discovery Unit, GlaxoSmithKline R&D, GSK R&D, Research Triangle Park, North Carolina, United States of America
| |
Collapse
|
1124
|
4-Benzofuranyloxynicotinamide derivatives are novel potent and orally available TGR5 agonists. Eur J Med Chem 2014; 82:1-15. [DOI: 10.1016/j.ejmech.2014.05.031] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Revised: 05/09/2014] [Accepted: 05/11/2014] [Indexed: 01/22/2023]
|
1125
|
Roda A, Pellicciari R, Gioiello A, Neri F, Camborata C, Passeri D, De Franco F, Spinozzi S, Colliva C, Adorini L, Montagnani M, Aldini R. Semisynthetic bile acid FXR and TGR5 agonists: physicochemical properties, pharmacokinetics, and metabolism in the rat. J Pharmacol Exp Ther 2014; 350:56-68. [PMID: 24784847 DOI: 10.1124/jpet.114.214650] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025] Open
Abstract
We report on the relationship between the structure-pharmacokinetics, metabolism, and therapeutic activity of semisynthetic bile acid analogs, including 6α-ethyl-3α,7α-dihydroxy-5β-cholan-24-oic acid (a selective farnesoid X receptor [FXR] receptor agonist), 6α-ethyl-23(S)-methyl-3α,7α,12α-trihydroxy-5β-cholan-24-oic acid (a specific Takeda G protein-coupled receptor 5 [TGR5] receptor agonist), and 6α-ethyl-3α,7α-dihydroxy-24-nor-5β-cholan-23-sulfate (a dual FXR/TGR5 agonist). We measured the main physicochemical properties of these molecules, including ionization constants, water solubility, lipophilicity, detergency, and protein binding. Biliary secretion and metabolism and plasma and hepatic concentrations were evaluated by high-pressure liquid chromatography-electrospray-mass spectrometry/mass spectrometry in bile fistula rat and compared with natural analogs chenodeoxycholic, cholic acid, and taurochenodexycholic acid and intestinal bacteria metabolism was evaluated in terms of 7α-dehydroxylase substrate-specificity in anaerobic human stool culture. The semisynthetic derivatives detergency, measured in terms of their critical micellar concentration, was quite similar to the natural analogs. They were slightly more lipophilic than the corresponding natural analogs, evaluated by their 1-octanol water partition coefficient (log P), because of the ethyl group in 6 position, which makes these molecules very stable toward bacterial 7-dehydroxylation. The hepatic metabolism and biliary secretion were different: 6α-ethyl-3α,7α-dihydroxy-5β-cholan-24-oic acid, as chenodeoxycholic acid, was efficiently conjugated with taurine in the liver and, only in this form, promptly and efficiently secreted in bile. 6α-Ethyl-23(S)-methyl-3α,7α,12α-trihydroxy-5β-cholan-24-oic acid was poorly conjugated with taurine because of the steric hindrance of the methyl at C23(S) position metabolized to the C23(R) isomer and partly conjugated with taurine. Conversely, 6α-ethyl-3α,7α-dihydroxy-24-nor-5β-cholan-23-sulfate was secreted in bile unmodified and as 3-glucuronide. Therefore, minor structural modifications profoundly influence the metabolism and biodistribution in the target organs where these analogs exert therapeutic effects by interacting with FXR and/or TGR5 receptors.
Collapse
Affiliation(s)
- Aldo Roda
- Dipartimento di Chimica "G. Ciamician" Alma Mater Studiorum-University of Bologna, Italy (A.R., C:C:, S.S.); Dipartimento di Chimica e Tecnologia del Farmaco, Università degli Studi di Perugia, Italy (R.P., A.G.); Dipartimento di Scienze Mediche e Chirurgiche, Alma Mater Studiorum-University of Bologna, Italy (F.N., M.M.); TES Pharma S.r.l., Corciano, Italy (R.P., D.P., F.D.F., C.C.); Intercept Pharmaceuticals, Inc., New York, New York; and Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum-University of Bologna, Bologna, Italy (R.A.)
| | - Roberto Pellicciari
- Dipartimento di Chimica "G. Ciamician" Alma Mater Studiorum-University of Bologna, Italy (A.R., C:C:, S.S.); Dipartimento di Chimica e Tecnologia del Farmaco, Università degli Studi di Perugia, Italy (R.P., A.G.); Dipartimento di Scienze Mediche e Chirurgiche, Alma Mater Studiorum-University of Bologna, Italy (F.N., M.M.); TES Pharma S.r.l., Corciano, Italy (R.P., D.P., F.D.F., C.C.); Intercept Pharmaceuticals, Inc., New York, New York; and Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum-University of Bologna, Bologna, Italy (R.A.)
| | - Antimo Gioiello
- Dipartimento di Chimica "G. Ciamician" Alma Mater Studiorum-University of Bologna, Italy (A.R., C:C:, S.S.); Dipartimento di Chimica e Tecnologia del Farmaco, Università degli Studi di Perugia, Italy (R.P., A.G.); Dipartimento di Scienze Mediche e Chirurgiche, Alma Mater Studiorum-University of Bologna, Italy (F.N., M.M.); TES Pharma S.r.l., Corciano, Italy (R.P., D.P., F.D.F., C.C.); Intercept Pharmaceuticals, Inc., New York, New York; and Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum-University of Bologna, Bologna, Italy (R.A.)
| | - Flavia Neri
- Dipartimento di Chimica "G. Ciamician" Alma Mater Studiorum-University of Bologna, Italy (A.R., C:C:, S.S.); Dipartimento di Chimica e Tecnologia del Farmaco, Università degli Studi di Perugia, Italy (R.P., A.G.); Dipartimento di Scienze Mediche e Chirurgiche, Alma Mater Studiorum-University of Bologna, Italy (F.N., M.M.); TES Pharma S.r.l., Corciano, Italy (R.P., D.P., F.D.F., C.C.); Intercept Pharmaceuticals, Inc., New York, New York; and Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum-University of Bologna, Bologna, Italy (R.A.)
| | - Cecilia Camborata
- Dipartimento di Chimica "G. Ciamician" Alma Mater Studiorum-University of Bologna, Italy (A.R., C:C:, S.S.); Dipartimento di Chimica e Tecnologia del Farmaco, Università degli Studi di Perugia, Italy (R.P., A.G.); Dipartimento di Scienze Mediche e Chirurgiche, Alma Mater Studiorum-University of Bologna, Italy (F.N., M.M.); TES Pharma S.r.l., Corciano, Italy (R.P., D.P., F.D.F., C.C.); Intercept Pharmaceuticals, Inc., New York, New York; and Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum-University of Bologna, Bologna, Italy (R.A.)
| | - Daniela Passeri
- Dipartimento di Chimica "G. Ciamician" Alma Mater Studiorum-University of Bologna, Italy (A.R., C:C:, S.S.); Dipartimento di Chimica e Tecnologia del Farmaco, Università degli Studi di Perugia, Italy (R.P., A.G.); Dipartimento di Scienze Mediche e Chirurgiche, Alma Mater Studiorum-University of Bologna, Italy (F.N., M.M.); TES Pharma S.r.l., Corciano, Italy (R.P., D.P., F.D.F., C.C.); Intercept Pharmaceuticals, Inc., New York, New York; and Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum-University of Bologna, Bologna, Italy (R.A.)
| | - Francesca De Franco
- Dipartimento di Chimica "G. Ciamician" Alma Mater Studiorum-University of Bologna, Italy (A.R., C:C:, S.S.); Dipartimento di Chimica e Tecnologia del Farmaco, Università degli Studi di Perugia, Italy (R.P., A.G.); Dipartimento di Scienze Mediche e Chirurgiche, Alma Mater Studiorum-University of Bologna, Italy (F.N., M.M.); TES Pharma S.r.l., Corciano, Italy (R.P., D.P., F.D.F., C.C.); Intercept Pharmaceuticals, Inc., New York, New York; and Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum-University of Bologna, Bologna, Italy (R.A.)
| | - Silvia Spinozzi
- Dipartimento di Chimica "G. Ciamician" Alma Mater Studiorum-University of Bologna, Italy (A.R., C:C:, S.S.); Dipartimento di Chimica e Tecnologia del Farmaco, Università degli Studi di Perugia, Italy (R.P., A.G.); Dipartimento di Scienze Mediche e Chirurgiche, Alma Mater Studiorum-University of Bologna, Italy (F.N., M.M.); TES Pharma S.r.l., Corciano, Italy (R.P., D.P., F.D.F., C.C.); Intercept Pharmaceuticals, Inc., New York, New York; and Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum-University of Bologna, Bologna, Italy (R.A.)
| | - Carolina Colliva
- Dipartimento di Chimica "G. Ciamician" Alma Mater Studiorum-University of Bologna, Italy (A.R., C:C:, S.S.); Dipartimento di Chimica e Tecnologia del Farmaco, Università degli Studi di Perugia, Italy (R.P., A.G.); Dipartimento di Scienze Mediche e Chirurgiche, Alma Mater Studiorum-University of Bologna, Italy (F.N., M.M.); TES Pharma S.r.l., Corciano, Italy (R.P., D.P., F.D.F., C.C.); Intercept Pharmaceuticals, Inc., New York, New York; and Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum-University of Bologna, Bologna, Italy (R.A.)
| | - Luciano Adorini
- Dipartimento di Chimica "G. Ciamician" Alma Mater Studiorum-University of Bologna, Italy (A.R., C:C:, S.S.); Dipartimento di Chimica e Tecnologia del Farmaco, Università degli Studi di Perugia, Italy (R.P., A.G.); Dipartimento di Scienze Mediche e Chirurgiche, Alma Mater Studiorum-University of Bologna, Italy (F.N., M.M.); TES Pharma S.r.l., Corciano, Italy (R.P., D.P., F.D.F., C.C.); Intercept Pharmaceuticals, Inc., New York, New York; and Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum-University of Bologna, Bologna, Italy (R.A.)
| | - Marco Montagnani
- Dipartimento di Chimica "G. Ciamician" Alma Mater Studiorum-University of Bologna, Italy (A.R., C:C:, S.S.); Dipartimento di Chimica e Tecnologia del Farmaco, Università degli Studi di Perugia, Italy (R.P., A.G.); Dipartimento di Scienze Mediche e Chirurgiche, Alma Mater Studiorum-University of Bologna, Italy (F.N., M.M.); TES Pharma S.r.l., Corciano, Italy (R.P., D.P., F.D.F., C.C.); Intercept Pharmaceuticals, Inc., New York, New York; and Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum-University of Bologna, Bologna, Italy (R.A.)
| | - Rita Aldini
- Dipartimento di Chimica "G. Ciamician" Alma Mater Studiorum-University of Bologna, Italy (A.R., C:C:, S.S.); Dipartimento di Chimica e Tecnologia del Farmaco, Università degli Studi di Perugia, Italy (R.P., A.G.); Dipartimento di Scienze Mediche e Chirurgiche, Alma Mater Studiorum-University of Bologna, Italy (F.N., M.M.); TES Pharma S.r.l., Corciano, Italy (R.P., D.P., F.D.F., C.C.); Intercept Pharmaceuticals, Inc., New York, New York; and Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum-University of Bologna, Bologna, Italy (R.A.)
| |
Collapse
|
1126
|
Abstract
The human gastrointestinal tract hosts a large number of microbial cells which exceed their mammalian counterparts by approximately 3-fold. The genes expressed by these microorganisms constitute the gut microbiome and may participate in diverse functions that are essential to the host, including digestion, regulation of energy metabolism, and modulation of inflammation and immunity. The gut microbiome can be modulated by dietary changes, antibiotic use, or disease. Different ailments have distinct associated microbiomes in which certain species or genes are present in different relative quantities. Thus, identifying specific disease-associated signatures in the microbiome as well as the factors that alter microbial populations and gene expression will lead to the development of new products such as prebiotics, probiotics, antimicrobials, live biotherapeutic products, or more traditional drugs to treat these disorders. Gained knowledge on the microbiome may result in molecular lab tests that may serve as personalized tools to guide the use of the aforementioned products and monitor interventional progress.
Collapse
Affiliation(s)
| | | | | | | | - Satya Prakash
- Micropharma Limited; Montreal, QC Canada; Biomedical Technology and Cell Therapy Research Laboratory; Department of Biomedical Engineering; Faculty of Medicine; McGill University; Montreal, QC Canada
| |
Collapse
|
1127
|
Diep TA, Madsen AN, Krogh-Hansen S, Al-Shahwani M, Al-Sabagh L, Holst B, Hansen HS. Dietary non-esterified oleic Acid decreases the jejunal levels of anorectic N-acylethanolamines. PLoS One 2014; 9:e100365. [PMID: 24959837 PMCID: PMC4068999 DOI: 10.1371/journal.pone.0100365] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 05/27/2014] [Indexed: 01/22/2023] Open
Abstract
Background and Aims Oleoylethanolamide and several other N-acylethanolamines (NAEs), e.g. linoleoylethanolamide and palmitoylethanolamide, have anorectic properties in rats, and prolonged intake of a high-fat diet decreases the levels of the anorectic NAEs in jejunum. Jejunal anorectic NAEs are thought to add to the control of food intake via activation of PPARalpha and the vagus nerve. The fat-induced decrease may explain part of the hyperphagic effect of high-fat diets. In the present study, we investigated 1) whether the reduced levels of anorectic NAEs were reversible in rats, 2) whether mice respond to dietary fat (olive oil) by reducing levels of anorectic NAEs, and 3) whether dietary non-esterified oleic acid also can decrease levels of anorectic NAEs in mice. We are searching for the fat sensor in the intestine, which mediates the decreased levels of anorectic NAEs. Methods Male rats and mice were fed diets high (45 energy% fat) in either triacylglycerol or free fatty acids for 7–14 days, and jejunal NAE and N-acylphosphatidylethanolamine (NAPE) levels were determined by liquid-chromatography mass spectrometry. Results In rats, reduced levels of anorectic NAEs could be reversed after 3 days from changing the diet from high-fat to chow. Corresponding NAPE levels tended to show the same changes. In mice, jejunal levels of anorectic NAEs were also reduced when fed a high-fat diet. In addition, we found that non-esterified oleic acid were also able to reduce levels of anorectic NAEs in mice. Conclusions These results suggest that the down-regulation of the jejunal level of anorectic NAEs by dietary fat is not restricted to rats, and that the fatty acid component oleic acid, in dietary olive oil may be sufficient to mediate this regulation. Thus, a fatty acid sensor may mediate this effect of dietary fat.
Collapse
Affiliation(s)
- Thi Ai Diep
- Department of Drug Design & Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Andreas N. Madsen
- Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sandra Krogh-Hansen
- Department of Drug Design & Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marwa Al-Shahwani
- Department of Drug Design & Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Laila Al-Sabagh
- Department of Drug Design & Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Birgitte Holst
- Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Harald S. Hansen
- Department of Drug Design & Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| |
Collapse
|
1128
|
Lipid-lowering Therapies, Glucose Control and Incident Diabetes: Evidence, Mechanisms and Clinical Implications. Cardiovasc Drugs Ther 2014; 28:361-77. [DOI: 10.1007/s10557-014-6534-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
1129
|
Tsuei J, Chau T, Mills D, Wan YJY. Bile acid dysregulation, gut dysbiosis, and gastrointestinal cancer. Exp Biol Med (Maywood) 2014; 239:1489-504. [PMID: 24951470 DOI: 10.1177/1535370214538743] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Because of increasingly widespread sedentary lifestyles and diets high in fat and sugar, the global diabetes and obesity epidemic continues to grow unabated. A substantial body of evidence has been accumulated which associates diabetes and obesity to dramatically higher risk of cancer development, particularly in the liver and gastrointestinal tract. Additionally, diabetic and obese individuals have been shown to suffer from dysregulation of bile acid (BA) homeostasis and dysbiosis of the intestinal microbiome. Abnormally elevated levels of cytotoxic secondary BAs and a pro-inflammatory shift in gut microbial profile have individually been linked to numerous enterohepatic diseases including cancer. However, recent findings have implicated a detrimental interplay between BA dysregulation and intestinal dysbiosis that promotes carcinogenesis along the gut-liver axis. This review seeks to examine the currently investigated interactions between the regulation of BA metabolism and activity of the intestinal microbiota and how these interactions can drive cancer formation in the context of diabesity. The precarcinogenic effects of BA dysregulation and gut dysbiosis including excessive inflammation, heightened oxidative DNA damage, and increased cell proliferation are discussed. Furthermore, by focusing on the mediatory roles of BA nuclear receptor farnesoid x receptor, ileal transporter apical sodium dependent BA transporter, and G-coupled protein receptor TGR5, this review attempts to connect BA dysregulation, gut dysbiosis, and enterohepatic carcinogenesis at a mechanistic level. A better understanding of the intricate interplay between BA homeostasis and gut microbiome can yield novel avenues to combat the impending rise in diabesity-related cancers.
Collapse
Affiliation(s)
- Jessica Tsuei
- Department of Pathology and Laboratory Medicine, University of California at Davis Medical Center, Sacramento, CA 95831, USA
| | - Thinh Chau
- Department of Pathology and Laboratory Medicine, University of California at Davis Medical Center, Sacramento, CA 95831, USA
| | - David Mills
- Department of Food Science and Technology, Department of Viticulture and Enology, Foods for Health Institute, University of California, Davis, CA 95616, USA
| | - Yu-Jui Yvonne Wan
- Department of Pathology and Laboratory Medicine, University of California at Davis Medical Center, Sacramento, CA 95831, USA
| |
Collapse
|
1130
|
Baghdasaryan A, Chiba P, Trauner M. Clinical application of transcriptional activators of bile salt transporters. Mol Aspects Med 2014; 37:57-76. [PMID: 24333169 PMCID: PMC4045202 DOI: 10.1016/j.mam.2013.12.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 11/21/2013] [Accepted: 12/01/2013] [Indexed: 02/07/2023]
Abstract
Hepatobiliary bile salt (BS) transporters are critical determinants of BS homeostasis controlling intracellular concentrations of BSs and their enterohepatic circulation. Genetic or acquired dysfunction of specific transport systems causes intrahepatic and systemic retention of potentially cytotoxic BSs, which, in high concentrations, may disturb integrity of cell membranes and subcellular organelles resulting in cell death, inflammation and fibrosis. Transcriptional regulation of canalicular BS efflux through bile salt export pump (BSEP), basolateral elimination through organic solute transporters alpha and beta (OSTα/OSTβ) as well as inhibition of hepatocellular BS uptake through basolateral Na(+)-taurocholate cotransporting polypeptide (NTCP) represent critical steps in protection from hepatocellular BS overload and can be targeted therapeutically. In this article, we review the potential clinical implications of the major BS transporters BSEP, OSTα/OSTβ and NTCP in the pathogenesis of hereditary and acquired cholestatic syndromes, provide an overview on transcriptional control of these transporters by the key regulatory nuclear receptors and discuss the potential therapeutic role of novel transcriptional activators of BS transporters in cholestasis.
Collapse
Affiliation(s)
- Anna Baghdasaryan
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Austria; Laboratory of Experimental and Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Austria
| | - Peter Chiba
- Institute of Medical Chemistry, Medical University of Vienna, Austria
| | - Michael Trauner
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Austria.
| |
Collapse
|
1131
|
Chatzigeorgiou A, Kandaraki E, Papavassiliou AG, Koutsilieris M. Peripheral targets in obesity treatment: a comprehensive update. Obes Rev 2014; 15:487-503. [PMID: 24612276 DOI: 10.1111/obr.12163] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Revised: 12/20/2013] [Accepted: 01/07/2014] [Indexed: 12/17/2022]
Abstract
Obesity is a major epidemic of our time and is associated with diseases such as metabolic syndrome, type 2 diabetes mellitus and atherosclerotic cardiovascular disease. Although weight loss drugs, when accompanied by diet and exercise, could be a very helpful medical tool in treating obese or overweight patients, their usefulness has been questioned due to the complexity of this type of medication, which regards a plethora of issues such as efficacy and safety of the drug and also risks and benefits among different patients. In general, obesity drugs that target peripheral pathophysiological mechanisms can be divided into two main categories. The first category includes anti-obesity agents able to reduce or limit energy absorption, such as pancreatic lipase and microsomal triglyceride transfer protein inhibitors. The second category consists of a heterogeneous group of compounds aiming to decrease fat mass by increasing energy expenditure or by redistributing adipose tissue. Angiogenesis inhibitors, beta-3 receptor agonists, sirtuin-I activators, diazoxide and other molecules belong to this group. The glucagon-like peptide-1 receptor agonists consist the third category of peripheral anti-obesity agents discussed therein. This review aims to provide a general overview of the molecules and substances that are already or could potentially be used as peripheral anti-obesity drugs, the molecular mechanisms by which they act, as well as their current stage of development, production and/or availability.
Collapse
Affiliation(s)
- A Chatzigeorgiou
- Department of Experimental Physiology, University of Athens Medical School, Athens, Greece; Department of Internal Medicine III and Institute of Physiology, University of Dresden, Dresden, Germany
| | | | | | | |
Collapse
|
1132
|
Abstract
Thyroid hormone (TH) is required for normal development as well as regulating metabolism in the adult. The thyroid hormone receptor (TR) isoforms, α and β, are differentially expressed in tissues and have distinct roles in TH signaling. Local activation of thyroxine (T4), to the active form, triiodothyronine (T3), by 5'-deiodinase type 2 (D2) is a key mechanism of TH regulation of metabolism. D2 is expressed in the hypothalamus, white fat, brown adipose tissue (BAT), and skeletal muscle and is required for adaptive thermogenesis. The thyroid gland is regulated by thyrotropin releasing hormone (TRH) and thyroid stimulating hormone (TSH). In addition to TRH/TSH regulation by TH feedback, there is central modulation by nutritional signals, such as leptin, as well as peptides regulating appetite. The nutrient status of the cell provides feedback on TH signaling pathways through epigentic modification of histones. Integration of TH signaling with the adrenergic nervous system occurs peripherally, in liver, white fat, and BAT, but also centrally, in the hypothalamus. TR regulates cholesterol and carbohydrate metabolism through direct actions on gene expression as well as cross-talk with other nuclear receptors, including peroxisome proliferator-activated receptor (PPAR), liver X receptor (LXR), and bile acid signaling pathways. TH modulates hepatic insulin sensitivity, especially important for the suppression of hepatic gluconeogenesis. The role of TH in regulating metabolic pathways has led to several new therapeutic targets for metabolic disorders. Understanding the mechanisms and interactions of the various TH signaling pathways in metabolism will improve our likelihood of identifying effective and selective targets.
Collapse
|
1133
|
Hofmann AF, Hagey LR. Key discoveries in bile acid chemistry and biology and their clinical applications: history of the last eight decades. J Lipid Res 2014; 55:1553-95. [PMID: 24838141 DOI: 10.1194/jlr.r049437] [Citation(s) in RCA: 246] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Indexed: 12/12/2022] Open
Abstract
During the last 80 years there have been extraordinary advances in our knowledge of the chemistry and biology of bile acids. We present here a brief history of the major achievements as we perceive them. Bernal, a physicist, determined the X-ray structure of cholesterol crystals, and his data together with the vast chemical studies of Wieland and Windaus enabled the correct structure of the steroid nucleus to be deduced. Today, C24 and C27 bile acids together with C27 bile alcohols constitute most of the bile acid "family". Patterns of bile acid hydroxylation and conjugation are summarized. Bile acid measurement encompasses the techniques of GC, HPLC, and MS, as well as enzymatic, bioluminescent, and competitive binding methods. The enterohepatic circulation of bile acids results from vectorial transport of bile acids by the ileal enterocyte and hepatocyte; the key transporters have been cloned. Bile acids are amphipathic, self-associate in solution, and form mixed micelles with polar lipids, phosphatidylcholine in bile, and fatty acids in intestinal content during triglyceride digestion. The rise and decline of dissolution of cholesterol gallstones by the ingestion of 3,7-dihydroxy bile acids is chronicled. Scientists from throughout the world have contributed to these achievements.
Collapse
Affiliation(s)
- Alan F Hofmann
- Department of Medicine, University of California, San Diego, San Diego, CA
| | - Lee R Hagey
- Department of Medicine, University of California, San Diego, San Diego, CA
| |
Collapse
|
1134
|
Qi Y, Jiang C, Cheng J, Krausz KW, Li T, Ferrell JM, Gonzalez FJ, Chiang JYL. Bile acid signaling in lipid metabolism: metabolomic and lipidomic analysis of lipid and bile acid markers linked to anti-obesity and anti-diabetes in mice. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1851:19-29. [PMID: 24796972 DOI: 10.1016/j.bbalip.2014.04.008] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 04/17/2014] [Accepted: 04/28/2014] [Indexed: 12/11/2022]
Abstract
Bile acid synthesis is the major pathway for catabolism of cholesterol. Cholesterol 7α-hydroxylase (CYP7A1) is the rate-limiting enzyme in the bile acid biosynthetic pathway in the liver and plays an important role in regulating lipid, glucose and energy metabolism. Transgenic mice overexpressing CYP7A1 (CYP7A1-tg mice) were resistant to high-fat diet (HFD)-induced obesity, fatty liver, and diabetes. However the mechanism of resistance to HFD-induced obesity of CYP7A1-tg mice has not been determined. In this study, metabolomic and lipidomic profiles of CYP7A1-tg mice were analyzed to explore the metabolic alterations in CYP7A1-tg mice that govern the protection against obesity and insulin resistance by using ultra-performance liquid chromatography-coupled with electrospray ionization quadrupole time-of-flight mass spectrometry combined with multivariate analyses. Lipidomics analysis identified seven lipid markers including lysophosphatidylcholines, phosphatidylcholines, sphingomyelins and ceramides that were significantly decreased in serum of HFD-fed CYP7A1-tg mice. Metabolomics analysis identified 13 metabolites in bile acid synthesis including taurochenodeoxycholic acid, taurodeoxycholic acid, tauroursodeoxycholic acid, taurocholic acid, and tauro-β-muricholic acid (T-β-MCA) that differed between CYP7A1-tg and wild-type mice. Notably, T-β-MCA, an antagonist of the farnesoid X receptor (FXR) was significantly increased in intestine of CYP7A1-tg mice. This study suggests that reducing 12α-hydroxylated bile acids and increasing intestinal T-β-MCA may reduce high fat diet-induced increase of phospholipids, sphingomyelins and ceramides, and ameliorate diabetes and obesity. This article is part of a Special Issue entitled Linking transcription to physiology in lipodomics.
Collapse
Affiliation(s)
- Yunpeng Qi
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Changtao Jiang
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jie Cheng
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Kristopher W Krausz
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tiangang Li
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Jessica M Ferrell
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - John Y L Chiang
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA.
| |
Collapse
|
1135
|
Nieuwdorp M, Gilijamse PW, Pai N, Kaplan LM. Role of the microbiome in energy regulation and metabolism. Gastroenterology 2014; 146:1525-33. [PMID: 24560870 DOI: 10.1053/j.gastro.2014.02.008] [Citation(s) in RCA: 294] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 01/25/2014] [Accepted: 02/16/2014] [Indexed: 02/06/2023]
Abstract
Intestinal microbes regulate metabolic function and energy balance; an altered microbial ecology is believed to contribute to the development of several metabolic diseases. Relative species abundance and metabolic characteristics of the intestinal microbiota change substantially in those who are obese or have other metabolic disorders and in response to ingested nutrients or therapeutic agents. The mechanisms through which the intestinal microbiota and its metabolites affect host homeostasis are just beginning to be understood. We review the relationships between the intestinal microbiota and host metabolism, including energy intake, use, and expenditure, in relation to glucose and lipid metabolism. These associations, along with interactions among the intestinal microbiota, mucus layer, bile acids, and mucosal immune responses, reveal potential mechanisms by which the microbiota affect metabolism. We discuss how controlled studies involving direct perturbations of microbial communities in human and animal models are required to identify effective therapeutic targets in the microbiota.
Collapse
Affiliation(s)
- Max Nieuwdorp
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Wallenberg Laboratory, Sahlgrenska Center for Cardiovascular and Metabolic Research, University of Göteborg, Göteborg, Sweden.
| | - Pim W Gilijamse
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Nikhil Pai
- Department of Pediatric Gastroenterology & Nutrition, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts; Obesity, Metabolism and Nutrition Institute and Gastrointestinal Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Lee M Kaplan
- Obesity, Metabolism and Nutrition Institute and Gastrointestinal Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
1136
|
Schnabl B, Brenner DA. Interactions between the intestinal microbiome and liver diseases. Gastroenterology 2014; 146:1513-24. [PMID: 24440671 PMCID: PMC3996054 DOI: 10.1053/j.gastro.2014.01.020] [Citation(s) in RCA: 743] [Impact Index Per Article: 67.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 12/29/2013] [Accepted: 01/07/2014] [Indexed: 02/08/2023]
Abstract
The human intestine harbors a diverse community of microbes that promote metabolism and digestion in their symbiotic relationship with the host. Disturbance of its homeostasis can result in disease. We review factors that disrupt intestinal homeostasis and contribute to nonalcoholic fatty liver disease, steatohepatitis, alcoholic liver disease, and cirrhosis. Liver disease has long been associated with qualitative and quantitative (overgrowth) dysbiotic changes in the intestinal microbiota. Extrinsic factors, such as the Western diet and alcohol, contribute to these changes. Dysbiosis results in intestinal inflammation, a breakdown of the intestinal barrier, and translocation of microbial products in animal models. However, the contribution of the intestinal microbiome to liver disease goes beyond simple translocation of bacterial products that promote hepatic injury and inflammation. Microbial metabolites produced in a dysbiotic intestinal environment and host factors are equally important in the pathogenesis of liver disease. We review how the combination of liver insult and disruptions in intestinal homeostasis contribute to liver disease.
Collapse
Affiliation(s)
- Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, California.
| | | |
Collapse
|
1137
|
Prawitt J, Caron S, Staels B. Glucose-lowering effects of intestinal bile acid sequestration through enhancement of splanchnic glucose utilization. Trends Endocrinol Metab 2014; 25:235-44. [PMID: 24731596 DOI: 10.1016/j.tem.2014.03.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 03/17/2014] [Accepted: 03/18/2014] [Indexed: 02/06/2023]
Abstract
Intestinal bile acid (BA) sequestration efficiently lowers plasma glucose concentrations in type 2 diabetes (T2D) patients. Because BAs act as signaling molecules via receptors, including the G protein-coupled receptor TGR5 and the nuclear receptor FXR (farnesoid X receptor), to regulate glucose homeostasis, BA sequestration, which interrupts the entero-hepatic circulation of BAs, constitutes a plausible action mechanism of BA sequestrants. An increase of intestinal L-cell glucagon-like peptide-1 (GLP-1) secretion upon TGR5 activation is the most commonly proposed mechanism, but recent studies also argue for a direct entero-hepatic action to enhance glucose utilization. We discuss here recent findings on the mechanisms of sequestrant-mediated glucose lowering via an increase of splanchnic glucose utilization through entero-hepatic FXR signaling.
Collapse
Affiliation(s)
- Janne Prawitt
- European Genomic Institute for Diabetes (EGID), FR 3508, 59000 Lille, France; Université Lille 2, 59000 Lille, France; Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR) 1011, 59000 Lille, France; Institut Pasteur de Lille, 59000 Lille, France
| | - Sandrine Caron
- European Genomic Institute for Diabetes (EGID), FR 3508, 59000 Lille, France; Université Lille 2, 59000 Lille, France; Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR) 1011, 59000 Lille, France; Institut Pasteur de Lille, 59000 Lille, France
| | - Bart Staels
- European Genomic Institute for Diabetes (EGID), FR 3508, 59000 Lille, France; Université Lille 2, 59000 Lille, France; Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR) 1011, 59000 Lille, France; Institut Pasteur de Lille, 59000 Lille, France.
| |
Collapse
|
1138
|
Lou G, Ma X, Fu X, Meng Z, Zhang W, Wang YD, Van Ness C, Yu D, Xu R, Huang W. GPBAR1/TGR5 mediates bile acid-induced cytokine expression in murine Kupffer cells. PLoS One 2014; 9:e93567. [PMID: 24755711 PMCID: PMC3995640 DOI: 10.1371/journal.pone.0093567] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 03/06/2014] [Indexed: 12/21/2022] Open
Abstract
GPBAR1/TGR5 is a novel plasma membrane-bound G protein–coupled bile acid (BA) receptor. BAs are known to induce the expression of inflammatory cytokines in the liver with unknown mechanism. Here we show that without other external stimuli, TGR5 activation alone induced the expression of interleukin 1β (IL-1β) and tumor necrosis factor-α (TNF-α) in murine macrophage cell line RAW264.7 or murine Kupffer cells. The TGR5-mediated increase of pro-inflammatory cytokine expression was suppressed by JNK inhibition. Moreover, the induced pro-inflammatory cytokine expression in mouse liver by 1% cholic acid (CA) diet was blunted in JNK−/− mice. TGR5 activation by its ligands enhanced the phosphorylation levels, DNA-binding and trans-activities of c-Jun and ATF2 transcription factors. Finally, the induced pro-inflammatory cytokine expression in Kupffer cells by TGR5 activation correlated with the suppression of Cholesterol 7α-hydroxylase (Cyp7a1) expression in murine hepatocytes. These results suggest that TGR5 mediates the BA-induced pro-inflammatory cytokine production in murine Kupffer cells through JNK-dependent pathway. This novel role of TGR5 may correlate to the suppression of Cyp7a1 expression in hepatocytes and contribute to the delicate BA feedback regulation.
Collapse
Affiliation(s)
- Guiyu Lou
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, China; Division of Molecular Diabetes Research, Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Xiaoxiao Ma
- Division of Molecular Diabetes Research, Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America; Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Xianghui Fu
- Division of Molecular Diabetes Research, Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Zhipeng Meng
- Division of Molecular Diabetes Research, Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America; Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Wenyu Zhang
- Division of Molecular Diabetes Research, Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Yan-Dong Wang
- Division of Molecular Diabetes Research, Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Carl Van Ness
- Division of Molecular Diabetes Research, Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Donna Yu
- Division of Molecular Diabetes Research, Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Rongzhen Xu
- Department of Hematology and Cancer Institute, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wendong Huang
- Division of Molecular Diabetes Research, Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| |
Collapse
|
1139
|
Taylor DR, Alaghband-Zadeh J, Cross GF, Omar S, le Roux CW, Vincent RP. Urine bile acids relate to glucose control in patients with type 2 diabetes mellitus and a body mass index below 30 kg/m2. PLoS One 2014; 9:e93540. [PMID: 24736330 PMCID: PMC3988028 DOI: 10.1371/journal.pone.0093540] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 03/04/2014] [Indexed: 01/06/2023] Open
Abstract
Bile acids are important endocrine signalling molecules, modulating glucose homeostasis through activation of cell surface and nuclear receptors. Bile acid metabolism is altered in type 2 diabetes mellitus; however, whether this is of pathogenic consequence is not fully established. In this study urinary bile acid excretion in individuals with type 2 diabetes and matched healthy volunteers was assessed. Urinary bile acid excretion in type 2 diabetes patients was considered in the context of prevailing glycaemia and the patient body mass index. Urine bile acids were measured by liquid chromatography-tandem mass spectrometry, allowing individual quantification of 15 bile acid species. Urinary bile acid excretion in patients with type 2 diabetes who were normal weight (BMI 18.5-24.9 kg/m2) and overweight (BMI 25-29.9 kg/m2) were elevated compared to healthy normal weight volunteers, both p<0.0001. In obese (BMI ≥ 30 kg/m2) type 2 diabetes patients, urinary bile acid excretion was significantly lower than in the normal and overweight type 2 diabetes groups (both p<0.01). Total bile acid excretion positively correlated with HbA1c in normal (rs=0.85, p=<0.001) and overweight (rs=0.61, p=0.02) but not obese type 2 diabetes patients (rs=-0.08, p=0.73). The glycaemia-associated increases in urine bile acid excretion in normal weight and overweight type 2 diabetes seen in this study may represent compensatory increases in bile acid signalling to maintain glucose homeostasis. As such alterations appear blunted by obesity; further investigation of weight-dependent effects of bile acid signalling on type 2 diabetes pathogenesis is warranted.
Collapse
Affiliation(s)
- David R. Taylor
- Department of Clinical Biochemistry, King's College Hospital, London, United Kingdom
| | | | - Gemma F. Cross
- Department of Clinical Biochemistry, King's College Hospital, London, United Kingdom
| | - Sohail Omar
- Department of Clinical Biochemistry, King's College Hospital, London, United Kingdom
| | - Carel W. le Roux
- Department of Clinical Biochemistry, King's College Hospital, London, United Kingdom
| | - Royce P. Vincent
- Department of Clinical Biochemistry, King's College Hospital, London, United Kingdom
| |
Collapse
|
1140
|
Phillips DP, Gao W, Yang Y, Zhang G, Lerario IK, Lau TL, Jiang J, Wang X, Nguyen DG, Bhat BG, Trotter C, Sullivan H, Welzel G, Landry J, Chen Y, Joseph SB, Li C, Gordon WP, Richmond W, Johnson K, Bretz A, Bursulaya B, Pan S, McNamara P, Seidel HM. Discovery of Trifluoromethyl(pyrimidin-2-yl)azetidine-2-carboxamides as Potent, Orally Bioavailable TGR5 (GPBAR1) Agonists: Structure–Activity Relationships, Lead Optimization, and Chronic In Vivo Efficacy. J Med Chem 2014; 57:3263-82. [DOI: 10.1021/jm401731q] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Dean P. Phillips
- Departments of †Medicinal
Chemistry, ‡Drug Discovery Biology, §Pharmacology, ∥Pharmacokinetics, and ⊥Structural Biology, Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Wenqi Gao
- Departments of †Medicinal
Chemistry, ‡Drug Discovery Biology, §Pharmacology, ∥Pharmacokinetics, and ⊥Structural Biology, Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Yang Yang
- Departments of †Medicinal
Chemistry, ‡Drug Discovery Biology, §Pharmacology, ∥Pharmacokinetics, and ⊥Structural Biology, Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Guobao Zhang
- Departments of †Medicinal
Chemistry, ‡Drug Discovery Biology, §Pharmacology, ∥Pharmacokinetics, and ⊥Structural Biology, Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Isabelle K. Lerario
- Departments of †Medicinal
Chemistry, ‡Drug Discovery Biology, §Pharmacology, ∥Pharmacokinetics, and ⊥Structural Biology, Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Thomas L. Lau
- Departments of †Medicinal
Chemistry, ‡Drug Discovery Biology, §Pharmacology, ∥Pharmacokinetics, and ⊥Structural Biology, Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Jiqing Jiang
- Departments of †Medicinal
Chemistry, ‡Drug Discovery Biology, §Pharmacology, ∥Pharmacokinetics, and ⊥Structural Biology, Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Xia Wang
- Departments of †Medicinal
Chemistry, ‡Drug Discovery Biology, §Pharmacology, ∥Pharmacokinetics, and ⊥Structural Biology, Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Deborah G. Nguyen
- Departments of †Medicinal
Chemistry, ‡Drug Discovery Biology, §Pharmacology, ∥Pharmacokinetics, and ⊥Structural Biology, Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - B. Ganesh Bhat
- Departments of †Medicinal
Chemistry, ‡Drug Discovery Biology, §Pharmacology, ∥Pharmacokinetics, and ⊥Structural Biology, Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Carol Trotter
- Departments of †Medicinal
Chemistry, ‡Drug Discovery Biology, §Pharmacology, ∥Pharmacokinetics, and ⊥Structural Biology, Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Heather Sullivan
- Departments of †Medicinal
Chemistry, ‡Drug Discovery Biology, §Pharmacology, ∥Pharmacokinetics, and ⊥Structural Biology, Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Gustav Welzel
- Departments of †Medicinal
Chemistry, ‡Drug Discovery Biology, §Pharmacology, ∥Pharmacokinetics, and ⊥Structural Biology, Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Jannine Landry
- Departments of †Medicinal
Chemistry, ‡Drug Discovery Biology, §Pharmacology, ∥Pharmacokinetics, and ⊥Structural Biology, Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Yali Chen
- Departments of †Medicinal
Chemistry, ‡Drug Discovery Biology, §Pharmacology, ∥Pharmacokinetics, and ⊥Structural Biology, Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Sean B. Joseph
- Departments of †Medicinal
Chemistry, ‡Drug Discovery Biology, §Pharmacology, ∥Pharmacokinetics, and ⊥Structural Biology, Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Chun Li
- Departments of †Medicinal
Chemistry, ‡Drug Discovery Biology, §Pharmacology, ∥Pharmacokinetics, and ⊥Structural Biology, Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - W. Perry Gordon
- Departments of †Medicinal
Chemistry, ‡Drug Discovery Biology, §Pharmacology, ∥Pharmacokinetics, and ⊥Structural Biology, Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Wendy Richmond
- Departments of †Medicinal
Chemistry, ‡Drug Discovery Biology, §Pharmacology, ∥Pharmacokinetics, and ⊥Structural Biology, Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Kevin Johnson
- Departments of †Medicinal
Chemistry, ‡Drug Discovery Biology, §Pharmacology, ∥Pharmacokinetics, and ⊥Structural Biology, Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Angela Bretz
- Departments of †Medicinal
Chemistry, ‡Drug Discovery Biology, §Pharmacology, ∥Pharmacokinetics, and ⊥Structural Biology, Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Badry Bursulaya
- Departments of †Medicinal
Chemistry, ‡Drug Discovery Biology, §Pharmacology, ∥Pharmacokinetics, and ⊥Structural Biology, Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Shifeng Pan
- Departments of †Medicinal
Chemistry, ‡Drug Discovery Biology, §Pharmacology, ∥Pharmacokinetics, and ⊥Structural Biology, Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Peter McNamara
- Departments of †Medicinal
Chemistry, ‡Drug Discovery Biology, §Pharmacology, ∥Pharmacokinetics, and ⊥Structural Biology, Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - H. Martin Seidel
- Departments of †Medicinal
Chemistry, ‡Drug Discovery Biology, §Pharmacology, ∥Pharmacokinetics, and ⊥Structural Biology, Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| |
Collapse
|
1141
|
Vrieze A, Out C, Fuentes S, Jonker L, Reuling I, Kootte RS, van Nood E, Holleman F, Knaapen M, Romijn JA, Soeters MR, Blaak EE, Dallinga-Thie GM, Reijnders D, Ackermans MT, Serlie MJ, Knop FK, Holst JJ, van der Ley C, Kema IP, Zoetendal EG, de Vos WM, Hoekstra JBL, Stroes ES, Groen AK, Nieuwdorp M. Impact of oral vancomycin on gut microbiota, bile acid metabolism, and insulin sensitivity. J Hepatol 2014; 60:824-31. [PMID: 24316517 DOI: 10.1016/j.jhep.2013.11.034] [Citation(s) in RCA: 424] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 11/21/2013] [Accepted: 11/25/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Obesity has been associated with changes in the composition and function of the intestinal microbiota. Modulation of the microbiota by antibiotics also alters bile acid and glucose metabolism in mice. Hence, we hypothesized that short term administration of oral antibiotics in humans would affect fecal microbiota composition and subsequently bile acid and glucose metabolism. METHODS In this single blinded randomized controlled trial, 20 male obese subjects with metabolic syndrome were randomized to 7 days of amoxicillin 500 mg t.i.d. or 7 days of vancomycin 500 mg t.i.d. At baseline and after 1 week of therapy, fecal microbiota composition (Human Intestinal Tract Chip phylogenetic microarray), fecal and plasma bile acid concentrations as well as insulin sensitivity (hyperinsulinemic euglycemic clamp using [6,6-(2)H2]-glucose tracer) were measured. RESULTS Vancomycin reduced fecal microbial diversity with a decrease of gram-positive bacteria (mainly Firmicutes) and a compensatory increase in gram-negative bacteria (mainly Proteobacteria). Concomitantly, vancomycin decreased fecal secondary bile acids with a simultaneous postprandial increase in primary bile acids in plasma (p<0.05). Moreover, changes in fecal bile acid concentrations were predominantly associated with altered Firmicutes. Finally, administration of vancomycin decreased peripheral insulin sensitivity (p<0.05). Amoxicillin did not affect any of these parameters. CONCLUSIONS Oral administration of vancomycin significantly impacts host physiology by decreasing intestinal microbiota diversity, bile acid dehydroxylation and peripheral insulin sensitivity in subjects with metabolic syndrome. These data show that intestinal microbiota, particularly of the Firmicutes phylum contributes to bile acid and glucose metabolism in humans. This trial is registered at the Dutch Trial Register (NTR2566).
Collapse
Affiliation(s)
- Anne Vrieze
- Department of Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Carolien Out
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Susana Fuentes
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Lisanne Jonker
- Department of Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Isaie Reuling
- Department of Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Ruud S Kootte
- Department of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Els van Nood
- Department of Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Frits Holleman
- Department of Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Max Knaapen
- Department of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Johannes A Romijn
- Department of Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Maarten R Soeters
- Department of Endocrinology and Metabolism, Academic Medical Center, Amsterdam, The Netherlands
| | - Ellen E Blaak
- Department of Human Metabolism, NUTRIM, School for Nutrition, Toxicology and Metabolism, Maastricht University, The Netherlands
| | - Geesje M Dallinga-Thie
- Department of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Dorien Reijnders
- Department of Human Metabolism, NUTRIM, School for Nutrition, Toxicology and Metabolism, Maastricht University, The Netherlands
| | - Mariëtte T Ackermans
- Department of Clinical Chemistry, Laboratory of Endocrinology, Academic Medical Center, Amsterdam, The Netherlands
| | - Mireille J Serlie
- Department of Endocrinology and Metabolism, Academic Medical Center, Amsterdam, The Netherlands
| | - Filip K Knop
- Department of Internal Medicine, Gentofte Hospital, Hellerup, Denmark; NNF Center for Basic Metabolic Research, Department of Biomedical Sciences, The Panum Institute, University of Copenhagen, Denmark
| | - Jenst J Holst
- NNF Center for Basic Metabolic Research, Department of Biomedical Sciences, The Panum Institute, University of Copenhagen, Denmark
| | - Claude van der Ley
- Department of Laboratory Medicine, University Medical Center Groningen, Groningen, The Netherlands
| | - Ido P Kema
- Department of Laboratory Medicine, University Medical Center Groningen, Groningen, The Netherlands
| | - Erwin G Zoetendal
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Willem M de Vos
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands; Department of Basic Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Joost B L Hoekstra
- Department of Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Erik S Stroes
- Department of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Albert K Groen
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Max Nieuwdorp
- Department of Medicine, Academic Medical Center, Amsterdam, The Netherlands; Department of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
1142
|
Wewalka M, Patti ME, Barbato C, Houten SM, Goldfine AB. Fasting serum taurine-conjugated bile acids are elevated in type 2 diabetes and do not change with intensification of insulin. J Clin Endocrinol Metab 2014; 99:1442-51. [PMID: 24432996 PMCID: PMC5393473 DOI: 10.1210/jc.2013-3367] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
CONTEXT Bile acids (BAs) are newly recognized signaling molecules in glucose and energy homeostasis. Differences in BA profiles with type 2 diabetes mellitus (T2D) remain incompletely understood. OBJECTIVE The objective of the study was to assess serum BA composition in impaired glucose-tolerant, T2D, and normal glucose-tolerant persons and to monitor the effects of improving glycemia on serum BA composition in T2D patients. DESIGN AND SETTING This was a cross-sectional cohort study in a general population (cohort 1) and nonrandomized intervention (cohort 2). PATIENTS AND INTERVENTIONS Ninety-nine volunteers underwent oral glucose tolerance testing, and 12 persons with T2D and hyperglycemia underwent 8 weeks of intensification of treatment. MAIN OUTCOME MEASURES Serum free BA and respective taurine and glycine conjugates were measured by HPLC tandem mass spectrometry. RESULTS Oral glucose tolerance testing identified 62 normal-, 25 impaired glucose-tolerant, and 12 T2D persons. Concentrations of total taurine-conjugated BA were higher in T2D and intermediate in impaired- compared with normal glucose-tolerant persons (P = .009). Univariate regression revealed a positive association between total taurine-BA and fasting glucose (R = 0.37, P < .001), postload glucose (R = 0.31, P < .002), hemoglobin A1c (R = 0.26, P < .001), fasting insulin (R = 0.21, P = .03), and homeostatic model assessment-estimated insulin resistance (R = 0.26, P = .01) and an inverse association with oral disposition index (R = -0.36, P < .001). Insulin-mediated glycemic improvement in T2D patients did not change fasting serum total BA or BA composition. CONCLUSION Fasting taurine-conjugated BA concentrations are higher in T2D and intermediate in impaired compared with normal glucose-tolerant persons and are associated with fasting and postload glucose. Serum BAs are not altered in T2D in response to improved glycemia. Further study may elucidate whether this pattern of taurine-BA conjugation can be targeted to provide novel therapeutic approaches to treat T2D.
Collapse
Affiliation(s)
- Marlene Wewalka
- Joslin Diabetes Center (M.W., M.-E.P., C.B., A.B.G.), Boston, Massachusetts 02215; Harvard Medical School (M.W., M.-E.P., A.B.G.), Harvard University, Boston, Massachusetts 02115; and Laboratory Genetic Metabolic Diseases (S.M.H.), Departments of Clinical Chemistry and Pediatrics, Academic Medical Center, 1105 AZ Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
1143
|
Li SK, Zhu D, Gaisano HY, Brubaker PL. Role of vesicle-associated membrane protein 2 in exocytosis of glucagon-like peptide-1 from the murine intestinal L cell. Diabetologia 2014; 57:809-18. [PMID: 24356748 DOI: 10.1007/s00125-013-3143-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 11/22/2013] [Indexed: 12/27/2022]
Abstract
AIMS/HYPOTHESIS Glucagon-like peptide-1 (GLP-1), secreted by the enteroendocrine L cell, is an incretin hormone that potently stimulates insulin secretion. Although signalling pathways promoting GLP-1 release are well characterised, the mechanisms by which GLP-1-containing granules fuse to the L cell membrane are unknown. As soluble NSF attachment proteins (SNAREs) are known to mediate granule-membrane fusion, the role of vesicle-associated membrane proteins (VAMPs) in GLP-1 exocytosis was examined. METHODS SNARE expression was determined in murine GLUTag L cells by RT-PCR and immunoblot and in primary murine L cells by immunofluorescence. Co-immunoprecipitation was used to examine SNARE interactions, while tetanus toxin (TetX)-mediated cleavage of VAMP was used with a GLP-1 secretion assay and total internal reflection fluorescence microscopy to determine the role of VAMP2 in exocytosis. RESULTS VAMP2 was expressed in murine L cells and localised to secretory granules in GLUTag cells. VAMP1/3 and the core membrane proteins syntaxin1a and synaptosomal-associated protein 25 kDa (SNAP25) were also detected. TetX cleaved VAMPs in GLUTag cells. However, only VAMP2 interacted with syntaxin1a, as did SNAP25 and Munc18-1. TetX treatment of GLUTag cells prevented glucose-dependent insulinotrophic peptide- and oleic-acid-stimulated GLP-1 secretion (p < 0.05-0.01), as well as K(+)-stimulated single-cell exocytosis (p < 0.05-0.001), while TetX-resistant VAMP2 expression rescued GLP-1 secretion (p < 0.01-0.001). CONCLUSIONS/INTERPRETATION Together, these findings indicate an essential role for VAMP2 in GLP-1 exocytosis from the GLUTag L cell in response to a variety of established secretagogues. An improved understanding of the mechanisms governing the release of GLP-1 may lead to new therapeutic approaches to enhance the levels of this incretin hormone in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Samantha K Li
- Department of Physiology, Medical Sciences Building, 1 King's College Circle, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | | | | | | |
Collapse
|
1144
|
Duboc H, Taché Y, Hofmann AF. The bile acid TGR5 membrane receptor: from basic research to clinical application. Dig Liver Dis 2014; 46:302-12. [PMID: 24411485 PMCID: PMC5953190 DOI: 10.1016/j.dld.2013.10.021] [Citation(s) in RCA: 345] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2013] [Revised: 10/03/2013] [Accepted: 10/31/2013] [Indexed: 02/07/2023]
Abstract
The TGR5 receptor (or GP-BAR1, or M-BAR) was characterized ten years ago as the first identified G-coupled protein receptor specific for bile acids. TGR5 gene expression is widely distributed, including endocrine glands, adipocytes, muscles, immune organs, spinal cord, and the enteric nervous system. The effect of TGR5 activation depends on the tissue where it is expressed and the signalling cascade that it induces. Animal studies suggest that TGR5 activation influences energy production and thereby may be involved in obesity and diabetes. TGR5 activation also influences intestinal motility. This review provides an overview of TGR5-bile acid interactions in health as well as the possible involvement of TGR5 in human disease.
Collapse
Affiliation(s)
- Henri Duboc
- Department of Medicine, CURE/Digestive Diseases Center and Center for Neurobiology of Stress, Digestive Diseases Division, University of California at Los Angeles, Los Angeles, CA, USA; Veterans Affairs Greater Los Angeles Healthcare System, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; University Paris Diderot, Sorbonne Paris Cité, AP-HP, Louis Mourier Hospital, Department of Gastroenterology and Hepatology, Paris, France; University Pierre and Marie Curie, ERL INSERM U 1057/UMR 7203, AP-HP, Saint-Antoine Hospital, Paris, France.
| | - Yvette Taché
- Department of Medicine, CURE/Digestive Diseases Center and Center for Neurobiology of Stress, Digestive Diseases Division, University of California at Los Angeles, Los Angeles, CA, USA; Veterans Affairs Greater Los Angeles Healthcare System, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Alan F Hofmann
- Division of Gastroenterology, Department of Medicine, University of California, San Diego, USA.
| |
Collapse
|
1145
|
Bunnett NW. Neuro-humoral signalling by bile acids and the TGR5 receptor in the gastrointestinal tract. J Physiol 2014; 592:2943-50. [PMID: 24614746 DOI: 10.1113/jphysiol.2014.271155] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In addition to their role in the digestion and absorption of dietary fats, bile acids (BAs) are tightly regulated signalling molecules. Their levels in the intestinal lumen, circulation and tissues fluctuate after feeding and fasting, and as a result of certain diseases and therapies. BAs regulate many cell types in the gut wall and beyond by activating nuclear and plasma membrane receptors. Of these, the G protein-coupled receptor TGR5 has emerged as a key mediator of the non-genomic actions of BAs. TGR5 is a cell-surface receptor that couples to Gαs, formation of cAMP, activation of protein kinase A and extracellular signal-regulated kinases, and inhibition of inflammatory signalling pathways. TGR5 has been implicated in mediating the actions of BAs on secretion of glucagon-like peptide 1 and glucose homeostasis, gastrointestinal motility and transit, electrolyte and fluid transport in the colon, bile formation and secretion, sensory transduction and inflammation. TGR5 agonists have been developed as treatments for metabolic, inflammatory and digestive disorders, and emerging evidence suggests that TGR5 mutations are associated with inflammatory diseases. Thus, TGR5 plays an important role in the normal processes of digestion and is a new therapeutic target for important digestive diseases.
Collapse
Affiliation(s)
- Nigel W Bunnett
- Monash Institute of Pharmaceutical Sciences, Parkville, Australia and Department of Pharmacology, University of Melbourne, Australia
| |
Collapse
|
1146
|
Kim K, Park M, Lee YM, Rhyu MR, Kim HY. Ginsenoside metabolite compound K stimulates glucagon-like peptide-1 secretion in NCI-H716 cells via bile acid receptor activation. Arch Pharm Res 2014; 37:1193-200. [PMID: 24590628 DOI: 10.1007/s12272-014-0362-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 02/19/2014] [Indexed: 12/13/2022]
Abstract
Compound K (CK) is a major metabolite of ginsenosides that is absorbed. CK has antidiabetic effects, although the mechanisms underlying the effects of CK have not fully been known. To elucidate the mechanisms underlying the antidiabetic effects of CK, we studied the effects of CK on GLP-1 secretion from NCI-H716 cells, and explored the mechanisms underlying CK-induced GLP-1 secretion. Treatment of NCI-H716 cells with 10, 50, and 100 μM CK significantly increased GLP-1 secretion, and intracellular Ca²⁺ and cAMP levels in a dose-dependent manner. Transfection of NCI-H716 cells with siRNA specific to α-gustducin and siRNA specific to TAS1R3 had no effect on CK-induced GLP-1 secretion and Ca²⁺ increase. However, transfection of NCI-H716 cells with TGR5-specific siRNA significantly inhibited CK-induced GLP-1 secretion and the increase in Ca²⁺ and cAMP levels. Moreover, CK showed human TGR5 agonist activity in CHO-K1 cells transiently transfected with human TGR5. Our data provide a novel mechanism of CK for antidiabetic effects. Moreover, the findings might suggest that CK is a potential agent that has multiple biological functions in the body via GLP-1 secretion and TGR5 activation.
Collapse
Affiliation(s)
- Kyong Kim
- Division of Metabolism and Functionality Research, Korea Food Research Institute, 62 Road-1201, Anyang-Pangyo-ro, Bundang-gu, Sungnam-si, Kyonggi-do, 463-746, Korea
| | | | | | | | | |
Collapse
|
1147
|
Poša M, Sebenji A. Determination of number-average aggregation numbers of bile salts micelles with a special emphasis on their oxo derivatives—The effect of the steroid skeleton. Biochim Biophys Acta Gen Subj 2014; 1840:1072-82. [DOI: 10.1016/j.bbagen.2013.11.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 10/27/2013] [Accepted: 11/07/2013] [Indexed: 01/22/2023]
|
1148
|
Liu Y, Hong T. Combination therapy of dipeptidyl peptidase-4 inhibitors and metformin in type 2 diabetes: rationale and evidence. Diabetes Obes Metab 2014; 16:111-7. [PMID: 23668534 DOI: 10.1111/dom.12128] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 02/25/2013] [Accepted: 05/06/2013] [Indexed: 01/22/2023]
Abstract
The main pathogenesis of type 2 diabetes mellitus (T2DM) includes insulin resistance and pancreatic islet dysfunction. Metformin, which attenuates insulin resistance, has been recommended as the first-line antidiabetic medication. Dipeptidyl peptidase-4 (DPP-4) inhibitors are novel oral hypoglycaemic agents that protect glucagon-like peptide-1 (GLP-1) from degradation, maintain the bioactivity of endogenous GLP-1, and thus improve islet dysfunction. Results from clinical trials have shown that the combination therapy of DPP-4 inhibitors and metformin [as an add-on, an initial combination or a fixed-dose combination (FDC)] provides excellent efficacy and safety in patients with T2DM. Moreover, recent studies have suggested that metformin enhances the biological effect of GLP-1 by increasing GLP-1 secretion, suppressing activity of DPP-4 and upregulating the expression of GLP-1 receptor in pancreatic β-cells. Conversely, DPP-4 inhibitors have a favourable effect on insulin sensitivity in patients with T2DM. Therefore, the combination of DPP-4 inhibitors and metformin provides an additive or even synergistic effect on metabolic control in patients with T2DM. This article provides an overview of clinical evidence and discusses the rationale for the combination therapy of DPP-4 inhibitors and metformin.
Collapse
Affiliation(s)
- Y Liu
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, China
| | | |
Collapse
|
1149
|
Jones ML, Martoni CJ, Ganopolsky JG, Labbé A, Prakash S. The human microbiome and bile acid metabolism: dysbiosis, dysmetabolism, disease and intervention. Expert Opin Biol Ther 2014; 14:467-82. [PMID: 24479734 DOI: 10.1517/14712598.2014.880420] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Recent evidence indicates that the human gut microbiome plays a significant role in health and disease. Dysbiosis, defined as a pathological imbalance in a microbial community, is becoming increasingly appreciated as a 'central environmental factor' that is both associated with complex phenotypes and affected by host genetics, diet and antibiotic use. More recently, a link has been established between the dysmetabolism of bile acids (BAs) in the gut to dysbiosis. AREAS COVERED BAs, which are transformed by the gut microbiota, have been shown to regulate intestinal homeostasis and are recognized as signaling molecules in a wide range of metabolic processes. This review will examine the connection between BA metabolism as it relates to the gut microbiome and its implication in health and disease. EXPERT OPINION A disrupted gut microbiome, including a reduction of bile salt hydrolase (BSH)-active bacteria, can significantly impair the metabolism of BAs and may result in an inability to maintain glucose homeostasis as well as normal cholesterol breakdown and excretion. To better understand the link between dysbiosis, BA dysmetabolism and chronic degenerative disease, large-scale metagenomic sequencing studies, metatranscriptomics, metaproteomics and metabolomics should continue to catalog functional diversity in the gastrointestinal tract of both healthy and diseased populations. Further, BSH-active probiotics should continue to be explored as treatment options to help restore metabolic levels.
Collapse
Affiliation(s)
- Mitchell L Jones
- McGill University, Artificial Cells and Organs Research Centre, Department of Biomedical Engineering and Physiology, Biomedical Technology and Cell Therapy Research Laboratory, Faculty of Medicine , 3775 University Street, Montreal, Quebec, H3A2B4 , Canada +1 514 398 3676 ; +1 514 398 7461 ;
| | | | | | | | | |
Collapse
|
1150
|
D’Amore C, Di Leva FS, Sepe V, Renga B, Del Gaudio C, D’Auria MV, Zampella A, Fiorucci S, Limongelli V. Design, Synthesis, and Biological Evaluation of Potent Dual Agonists of Nuclear and Membrane Bile Acid Receptors. J Med Chem 2014; 57:937-54. [DOI: 10.1021/jm401873d] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Claudio D’Amore
- Dipartimento
di Medicina Clinica e Sperimentale, Nuova Facoltà di Medicina, Università degli Studi di Perugia, Via Gambuli, 1-06132 Perugia, Italy
| | | | - Valentina Sepe
- Dipartimento
di Farmacia, Università di Napoli “Federico II”, Via D. Montesano, 49, I-80131 Napoli, Italy
| | - Barbara Renga
- Dipartimento
di Medicina Clinica e Sperimentale, Nuova Facoltà di Medicina, Università degli Studi di Perugia, Via Gambuli, 1-06132 Perugia, Italy
| | - Chiara Del Gaudio
- Dipartimento
di Farmacia, Università di Napoli “Federico II”, Via D. Montesano, 49, I-80131 Napoli, Italy
| | - Maria Valeria D’Auria
- Dipartimento
di Farmacia, Università di Napoli “Federico II”, Via D. Montesano, 49, I-80131 Napoli, Italy
| | - Angela Zampella
- Dipartimento
di Farmacia, Università di Napoli “Federico II”, Via D. Montesano, 49, I-80131 Napoli, Italy
| | - Stefano Fiorucci
- Dipartimento
di Medicina Clinica e Sperimentale, Nuova Facoltà di Medicina, Università degli Studi di Perugia, Via Gambuli, 1-06132 Perugia, Italy
| | - Vittorio Limongelli
- Dipartimento
di Farmacia, Università di Napoli “Federico II”, Via D. Montesano, 49, I-80131 Napoli, Italy
| |
Collapse
|