951
|
Abstract
Bile acids have many activities over and above their primary function in aiding absorption of fat and fat soluble vitamins. Bile acids are synthesized from cholesterol, and thus are involved in cholesterol homeostasis. Bile acids stimulate glucagon-like peptide 1 (GLP1) production in the distal small bowel and colon, stimulating insulin secretion, and therefore, are involved in carbohydrate and fat metabolism. Bile acids through their insulin sensitising effect play a part in insulin resistance and type 2 diabetes. Bile acid metabolism is altered in obesity and diabetes. Both dietary restriction and weight loss due to bariatric surgery, alter the lipid carbohydrate and bile acid metabolism. Recent research suggests that the forkhead transcription factor FOXO is a central regulator of bile, lipid, and carbohydrate metabolism, but conflicting studies mean that our understanding of the complexity is not yet complete.
Collapse
Affiliation(s)
- Gerald H Tomkin
- Diabetes Institute of Ireland Beacon Clinic Dublin and Trinity College, Dublin 2, Ireland
| | - Daphne Owens
- Diabetes Institute of Ireland Beacon Clinic Dublin and Trinity College, Dublin 2, Ireland
| |
Collapse
|
952
|
Finamore C, Festa C, Renga B, Sepe V, Carino A, Masullo D, Biagioli M, Marchianò S, Capolupo A, Monti MC, Fiorucci S, Zampella A. Navigation in bile acid chemical space: discovery of novel FXR and GPBAR1 ligands. Sci Rep 2016; 6:29320. [PMID: 27381677 PMCID: PMC4933954 DOI: 10.1038/srep29320] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 06/16/2016] [Indexed: 02/07/2023] Open
Abstract
Bile acids are signaling molecules interacting with nuclear receptors and membrane G-protein-coupled receptors. Among these receptors, the farnesoid X receptor (FXR) and the membrane G-coupled receptor (GPBAR1) have gained increasing consideration as druggable receptors and their exogenous dual regulation represents an attractive strategy in the treatment of enterohepatic and metabolic disorders. However, the therapeutic use of dual modulators could be associated to severe side effects and therefore the discovery of selective GPBAR1 and FXR agonists is an essential step in the medicinal chemistry optimization of bile acid scaffold. In this study, a new series of 6-ethylcholane derivatives modified on the tetracyclic core and on the side chain has been designed and synthesized and their in vitro activities on FXR and GPBAR1 were assayed. This speculation resulted in the identification of compound 7 as a potent and selective GPBAR1 agonist and of several derivatives showing potent dual agonistic activity.
Collapse
Affiliation(s)
- Claudia Finamore
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, 80131 Naples, Italy
| | - Carmen Festa
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, 80131 Naples, Italy
| | - Barbara Renga
- Department of Surgery and Biomedical Sciences, Nuova Facoltà di Medicina, P.zza L. Severi 1, 06132 Perugia, Italy
| | - Valentina Sepe
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, 80131 Naples, Italy
| | - Adriana Carino
- Department of Surgery and Biomedical Sciences, Nuova Facoltà di Medicina, P.zza L. Severi 1, 06132 Perugia, Italy
| | - Dario Masullo
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, 80131 Naples, Italy
| | - Michele Biagioli
- Department of Surgery and Biomedical Sciences, Nuova Facoltà di Medicina, P.zza L. Severi 1, 06132 Perugia, Italy
| | - Silvia Marchianò
- Department of Surgery and Biomedical Sciences, Nuova Facoltà di Medicina, P.zza L. Severi 1, 06132 Perugia, Italy
| | - Angela Capolupo
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano (Salerno), Italy
| | - Maria Chiara Monti
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano (Salerno), Italy
| | - Stefano Fiorucci
- Department of Surgery and Biomedical Sciences, Nuova Facoltà di Medicina, P.zza L. Severi 1, 06132 Perugia, Italy
| | - Angela Zampella
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, 80131 Naples, Italy
| |
Collapse
|
953
|
Zhang X, Liu T, Wang Y, Zhong M, Zhang G, Liu S, Wu T, Rayner CK, Hu S. Comparative Effects of Bile Diversion and Duodenal-Jejunal Bypass on Glucose and Lipid Metabolism in Male Diabetic Rats. Obes Surg 2016; 26:1565-1575. [PMID: 26464241 DOI: 10.1007/s11695-015-1925-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Duodenal-jejunal bypass (DJB) induces rapid and durable improvement in glucose and lipid metabolism. Besides bypassing the proximal gut, DJB also diverts bile acids (BAs) into the distal gut, increasing luminal and systemic BAs that are essential to metabolic homeostasis. The aim of this study is to evaluate whether bile diversion (BD) alone can recapitulate the effects of DJB on glucose and lipid metabolism. METHODS BD, DJB and SHAM procedures were performed in a diabetic rat model induced by high-fat diet (HFD)/streptozotocin (STZ). Body weight, energy intake, blood glucose, serum hormones, insulin sensitivity, lipid profiles, and luminal and systemic BAs were measured postsurgery. RESULTS BD reduced body weight, independently of energy intake, whereas DJB had no effects. Luminal and serum BAs were increased after both DJB and BD, and were higher after BD than DJB. During glucose tolerance test, both fasting and postprandial blood glucose concentrations were reduced with DJB and BD, and were lower after DJB than BD. Insulin sensitivity was improved after DJB, but remained unchanged after BD. Fasting and postprandial GLP-1 were equally increased after DJB and BD. Serum triglyceride and free fatty acids were decreased more after BD than DJB, while hepatic triglyceride storage was reduced more after DJB. CONCLUSION These observations indicate that BD, which increases luminal and systemic BAs and postprandial GLP-1, represents an important component of DJB in restoring glucose and lipid homeostasis in diabetic state. However, other mechanisms associated with DJB also appear to make complementary contributions to metabolic regulation.
Collapse
Affiliation(s)
- Xiang Zhang
- Department of General Surgery, Qilu Hospital of Shandong University, 107#, Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
- Discipline of Medicine, The University of Adelaide, Level 6 Eleanor Harrald Building, Royal Adelaide Hospital, Frome Road, Adelaide, SA, Australia
| | - Teng Liu
- Department of General Surgery, Qilu Hospital of Shandong University, 107#, Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Yanmin Wang
- Department of General Surgery, Qilu Hospital of Shandong University, 107#, Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Mingwei Zhong
- Department of General Surgery, Qilu Hospital of Shandong University, 107#, Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Guangyong Zhang
- Department of General Surgery, Qilu Hospital of Shandong University, 107#, Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Shaozhuang Liu
- Department of General Surgery, Qilu Hospital of Shandong University, 107#, Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Tongzhi Wu
- Discipline of Medicine, The University of Adelaide, Level 6 Eleanor Harrald Building, Royal Adelaide Hospital, Frome Road, Adelaide, SA, Australia
| | - Christopher K Rayner
- Discipline of Medicine, The University of Adelaide, Level 6 Eleanor Harrald Building, Royal Adelaide Hospital, Frome Road, Adelaide, SA, Australia
| | - Sanyuan Hu
- Department of General Surgery, Qilu Hospital of Shandong University, 107#, Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China.
| |
Collapse
|
954
|
Effects of various gastrointestinal procedures on β-cell function in obesity and type 2 diabetes. Surg Obes Relat Dis 2016; 12:1213-9. [DOI: 10.1016/j.soard.2016.02.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 02/23/2016] [Indexed: 12/18/2022]
|
955
|
Nunez DJ, Yao X, Lin J, Walker A, Zuo P, Webster L, Krug-Gourley S, Zamek-Gliszczynski MJ, Gillmor DS, Johnson SL. Glucose and lipid effects of the ileal apical sodium-dependent bile acid transporter inhibitor GSK2330672: double-blind randomized trials with type 2 diabetes subjects taking metformin. Diabetes Obes Metab 2016; 18:654-62. [PMID: 26939572 DOI: 10.1111/dom.12656] [Citation(s) in RCA: 26] [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: 12/13/2015] [Revised: 01/10/2016] [Accepted: 02/27/2016] [Indexed: 12/15/2022]
Abstract
AIMS To investigate the pharmacodynamics, pharmacokinetics and safety/tolerability of blocking reuptake of bile acids using the inhibitor GSK2330672 (GSK672) in patients with type 2 diabetes (T2D). METHODS Subjects with T2D taking metformin were enrolled in two studies in which they took metformin 850 mg twice daily for 2 weeks prior to and during the randomized treatment periods. In the first crossover study (n = 15), subjects received GSK672 45 mg, escalating to 90 mg, twice daily, or placebo for 7 days. The second parallel-group study (n = 75) investigated GSK672 10-90 mg twice daily, placebo or sitagliptin for 14 days. RESULTS In both studies, GSK672 reduced circulating bile acids and increased serum 7-α-hydroxy-4-cholesten-3-one (C4), an intermediate in the hepatic synthesis of bile acids. Compared with placebo, in the parallel-group study 90 mg GSK672 twice daily reduced fasting plasma glucose [FPG; -1.21 mmol/l; 95% confidence interval (CI) -2.14, -0.28] and weighted-mean glucose area under the curve (AUC)0-24 h (-1.33 mmol/l; 95% CI -2.30, -0.36), as well as fasting and weighted-mean insulin AUC0 -24 h . GSK672 also reduced cholesterol (LDL, non-HDL and total cholesterol) and apolipoprotein B concentrations; the maximum LDL cholesterol reduction was ∼40%. There was no change in HDL cholesterol but there was a trend towards increased fasting triglyceride levels in the GSK672 groups compared with placebo. In both studies, the most common adverse events associated with GSK672 were gastrointestinal, mostly diarrhoea (22-100%), which appeared to be independent of dose. CONCLUSIONS In subjects with T2D on metformin, GSK672 improved glucose and lipids, but there was a high incidence of gastrointestinal adverse events.
Collapse
Affiliation(s)
- D J Nunez
- GlaxoSmithKline plc, Research Triangle Park, NC and Collegeville, PA, USA
| | - X Yao
- Alexion Pharmaceuticals, Inc., Cambridge, MA, USA
| | - J Lin
- Grifols Therapeutics Inc., Research Triangle Park, NC, USA
| | - A Walker
- GlaxoSmithKline plc, Research Triangle Park, NC and Collegeville, PA, USA
| | - P Zuo
- Parexel International, Durham, NC, USA
| | | | - S Krug-Gourley
- GlaxoSmithKline plc, Research Triangle Park, NC and Collegeville, PA, USA
| | | | - D S Gillmor
- Pharmaceutical Product Development LLC, Morrisville, NC, USA
| | | |
Collapse
|
956
|
Intestinally-targeted TGR5 agonists equipped with quaternary ammonium have an improved hypoglycemic effect and reduced gallbladder filling effect. Sci Rep 2016; 6:28676. [PMID: 27339735 PMCID: PMC4919643 DOI: 10.1038/srep28676] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 06/06/2016] [Indexed: 12/31/2022] Open
Abstract
TGR5 activation of enteroendocrine cells increases glucagon-like peptide 1 (GLP-1) release, which maintains glycemic homeostasis. However, TGR5 activation in the gallbladder and heart is associated with severe side effects. Therefore, intestinally-targeted TGR5 agonists were suggested as potential hypoglycemic agents with minimal side effects. However, until now no such compounds with robust glucose-lowering effects were reported, especially in diabetic animal models. Herein, we identify a TGR5 agonist, 26a, which was proven to be intestinally-targeted through pharmacokinetic studies. 26a was used as a tool drug to verify the intestinally-targeted strategy. 26a displayed a robust and long-lasting hypoglycemic effect in ob/ob mice (once a day dosing (QD) and 18-day treatment) owing to sustained stimulation of GLP-1 secretion, which suggested that robust hypoglycemic effect could be achieved with activation of TGR5 in intestine alone. However, the gallbladder filling effect of 26a was rather complicated. Although the gallbladder filling effect of 26a was decreased in mice after once a day dosing, this side effect was still not eliminated. To solve the problem above, several research strategies were raised for further optimization.
Collapse
|
957
|
Bashiardes S, Shapiro H, Rozin S, Shibolet O, Elinav E. Non-alcoholic fatty liver and the gut microbiota. Mol Metab 2016; 5:782-94. [PMID: 27617201 PMCID: PMC5004228 DOI: 10.1016/j.molmet.2016.06.003] [Citation(s) in RCA: 180] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 06/04/2016] [Accepted: 06/08/2016] [Indexed: 02/06/2023] Open
Abstract
Background Non-alcoholic fatty liver (NAFLD) is a common, multi-factorial, and poorly understood liver disease whose incidence is globally rising. NAFLD is generally asymptomatic and associated with other manifestations of the metabolic syndrome. Yet, up to 25% of NAFLD patients develop a progressive inflammatory liver disease termed non-alcoholic steatohepatitis (NASH) that may progress towards cirrhosis, hepatocellular carcinoma, and the need for liver transplantation. In recent years, several lines of evidence suggest that the gut microbiome represents a significant environmental factor contributing to NAFLD development and its progression into NASH. Suggested microbiome-associated mechanisms contributing to NAFLD and NASH include dysbiosis-induced deregulation of the gut endothelial barrier function, which facilitates systemic bacterial translocation, and intestinal and hepatic inflammation. Furthermore, increased microbiome-modulated metabolites such as lipopolysaccharides, short chain fatty acids (SCFAs), bile acids, and ethanol, may affect liver pathology through multiple direct and indirect mechanisms. Scope of review Herein, we discuss the associations, mechanisms, and clinical implications of the microbiome's contribution to NAFLD and NASH. Understanding these contributions to the development of fatty liver pathogenesis and its clinical course may serve as a basis for development of therapeutic microbiome-targeting approaches for treatment and prevention of NAFLD and NASH. Major conclusions Intestinal host–microbiome interactions play diverse roles in the pathogenesis and progression of NAFLD and NASH. Elucidation of the mechanisms driving these microbial effects on the pathogenesis of NAFLD and NASH may enable to identify new diagnostic and therapeutic targets of these common metabolic liver diseases. This article is part of a special issue on microbiota.
Collapse
Affiliation(s)
- Stavros Bashiardes
- Immunology Department, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Hagit Shapiro
- Immunology Department, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Shachar Rozin
- Immunology Department, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Oren Shibolet
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel; Department of Gastroenterology, Tel Aviv Medical Center, Tel Aviv 6423906, Israel
| | - Eran Elinav
- Immunology Department, Weizmann Institute of Science, 76100 Rehovot, Israel
| |
Collapse
|
958
|
Abstract
Obesity and its associated medical conditions continue to increase and add significant burden to patients, as well as health-care systems, worldwide. Bariatric surgery is the most effective treatment for severe obesity and its comorbidities, and resolution of diabetes is weight loss-independent in the case of some operations. Although these weight-independent effects are frequently described clinically, the mechanisms behind them are not well understood and remain an intense area of focus in the growing field of metabolic and bariatric surgery. Perceptions of the mechanisms responsible for the beneficial metabolic effects of metabolic/bariatric operations have shifted from being mostly restrictive and malabsorption over the last 10 to 15 years to being more neuro-hormonal in origin. In this review, we describe recent basic and clinical findings of the major clinical procedures (adjustable gastric banding, vertical sleeve gastrectomy, Roux-en-Y gastric bypass, and biliopancreatic diversion) as well as other experimental procedures (ileal interposition and bile diversion) that recapitulate many of the metabolic effects of these complex operations in a simpler fashion. As the role of bile acids and the gut microbiome on metabolism is becoming increasingly well described, their potential roles in these improvements following metabolic surgery are becoming better appreciated. Bile acid and gut microbiome changes, in light of recent developments, are discussed in the context of these surgical procedures, as well as their implications for future study.
Collapse
Affiliation(s)
- Vance L Albaugh
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, USA
| | - C Robb Flynn
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, USA
| | - Robyn A Tamboli
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, USA
| | - Naji N Abumrad
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, USA
| |
Collapse
|
959
|
The effect of a tertiary bile acid, taurocholic acid, on the morphology and physical characteristics of microencapsulated probucol: potential applications in diabetes: a characterization study. Drug Deliv Transl Res 2016; 5:511-22. [PMID: 26242686 DOI: 10.1007/s13346-015-0248-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In recent studies, we designed multi-compartmental microcapsules as a platform for the targeted oral delivery of lipophilic drugs in an animal model of type 2 diabetes (T2D). Probucol (PB) is a highly lipophilic, antihyperlipidemic drug with potential antidiabetic effects. PB has low bioavailability and high inter-individual variations in absorption, which limits its clinical applications. In a new study, the bile acid, taurocholic acid (TCA), exerted permeation enhancing effects in vivo. Accordingly, this study aimed to design and characterize TCA-based PB microcapsules and examine the effects of TCA on the microcapsules' morphology, stability, and release profiles. Microcapsules were prepared using the polymer sodium alginate (SA). Two types of microcapsules were produced, one without TCA (PB-SA, control) and one with TCA (PB-TCA-SA, test). Microcapsules were studied in terms of morphology, surface structure and composition, size, drug contents, cross-sectional imaging (using microtomography (Micro-CT) analysis), Zeta potential, thermal and chemical profiles, rheological parameters, swelling, mechanical strength, and release studies at various temperature and pH values. The production yield and the encapsulation efficiency were also studied together with in vitro efficacy testing of cell viability at various glucose concentrations and insulin and TNF-α production using clonal-mouse pancreatic β-cells. PB-TCA-SA microcapsules showed uniform structure and even distribution of TCA within the microcapsules. Drug contents, Zeta potential, size, rheological parameters, production yield, and the microencapsulation efficiency remained similar after TCA addition. In vitro testing showed PB-TCA-SA microcapsules improved β-cell survival under hyperglycemic states and reduced the pro-inflammatory cytokine TNF-α while increasing insulin secretions compared with PB-SA microcapsules. PB-TCA-SA microcapsules also showed good stability, better mechanical (p < 0.01) and swelling (p < 0.01) characteristics, and optimized controlled release at pH 7.8 (p < 0.01) compared with control, suggesting desirable targeted release properties and potential applications in the oral delivery of PB in T2D.
Collapse
|
960
|
Expedited Biliopancreatic Juice Flow to the Distal Gut Benefits the Diabetes Control After Duodenal-Jejunal Bypass. Obes Surg 2016; 25:1802-9. [PMID: 25726319 DOI: 10.1007/s11695-015-1633-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Serum bile acids (BAs) are elevated after metabolic surgeries including Roux-en-Y gastric bypass (RYGB), ileal transposition (IT), and duodenal-jejunal bypass (DJB). Recently, BAs have emerged as a kind of signaling molecules, which can not only promote glucagon-like peptide-1 (GLP-1) secretion but can also regulate multiple enzymes involved in glucose metabolism. The aim of this study was to investigate whether expedited biliopancreatic juice flow to the distal gut contributes to the increased serum GLP-1 and BAs and benefits the diabetes control after DJB. METHODS DJB, long alimentary limb DJB (LDJB), duodenal-jejunal anastomosis (DJA), and sham operation were performed in diabetic rats induced by high-fat diet (HFD) and low dose of streptozotocin (STZ). Body weight, food intake, oral glucose tolerance, insulin tolerance, glucose-stimulated insulin and GLP-1 secretion, fasting serum total bile acids (TBAs), and lipid profiles were measured at indicated time points. RESULTS Compared with sham operation, DJA, DJB, and LDJB all achieved rapid and dramatic improvements in glucose tolerance and insulin sensitivity independently of food restriction and weight loss. DJB and LDJB-operated rats exhibited even better glucose tolerance, higher fasting serum TBAs, and higher glucose-stimulated GLP-1 secretion than the DJA group postoperatively. No difference was detected in insulin sensitivity and glucose-stimulated insulin secretion between DJA, DJB, and LDJB groups. CONCLUSIONS Expedited biliopancreatic juice flow to the distal gut was associated with augmented GLP-1 secretion and increased fasting serum TBA concentration, which may partly explain the metabolic benefits of DJB.
Collapse
|
961
|
Impact of gut microbiota on diabetes mellitus. DIABETES & METABOLISM 2016; 42:303-315. [PMID: 27179626 DOI: 10.1016/j.diabet.2016.04.004] [Citation(s) in RCA: 168] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 04/04/2016] [Accepted: 04/07/2016] [Indexed: 02/07/2023]
Abstract
Various functions of the gut are regulated by sophisticated interactions among its functional elements, including the gut microbiota. These microorganisms play a crucial role in gastrointestinal mucosa permeability. They control the fermentation and absorption of dietary polysaccharides to produce short-chain fatty acids, which may explain their importance in the regulation of fat accumulation and the subsequent development of obesity-related diseases, suggesting that they are a crucial mediator of obesity and its consequences. In addition, gut bacteria play a crucial role in the host immune system, modulation of inflammatory processes, extraction of energy from the host diet and alterations of human gene expression. Dietary modulation of the human colonic microbiota has been shown to confer a number of health benefits to the host. Simple therapeutic strategies targeted at attenuating the progression of chronic low-grade inflammation and insulin resistance are urgently required to prevent or slow the development of diabetes in susceptible individuals. The main objective of this review is to address the pathogenic association between gut microbiota and diabetes, and to explore any novel related therapeutic targets. New insights into the role of the gut microbiota in diabetes could lead to the development of integrated strategies using probiotics to prevent and treat these metabolic disorders.
Collapse
|
962
|
Krattinger R, Boström A, Lee SML, Thasler WE, Schiöth HB, Kullak-Ublick GA, Mwinyi J. Chenodeoxycholic acid significantly impacts the expression of miRNAs and genes involved in lipid, bile acid and drug metabolism in human hepatocytes. Life Sci 2016; 156:47-56. [PMID: 27174168 DOI: 10.1016/j.lfs.2016.04.037] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/21/2016] [Accepted: 04/27/2016] [Indexed: 02/07/2023]
Abstract
AIMS Bile acids (BAs) are important gut signaling hormones, influencing lipid, glucose, and energy homeostasis. The exact mechanisms behind these effects are not yet fully understood. Lately, they have come to the fore as putative therapeutics in metabolic diseases, such as e.g. nonalcoholic fatty liver disease (NAFLD). We elucidate to what extent BAs impacts on the mRNAome and microRNAome in hepatocytes to gather novel insights into the mechanisms behind metabolic and toxicologic effects of bile acids. MAIN METHODS Five batches of primary human hepatocytes were treated with 50μmol/l chenodeoxycholic acid (CDCA) for 24 or 48h. Total RNA was extracted, size fractionated and subjected to Next Generation Sequencing to generate mRNA and miRNA profiles. KEY FINDINGS Expression of 738 genes and 52 miRNAs were CDCA dependently decreased, whereas 1566 genes and 29 miRNAs were significantly increased in hepatocytes. Distinct gene clusters controlling BA and lipid homeostasis (FGF(R), APO and FABP family members, HMGCS2) and drug metabolism (CYP, UGT and SULT family members) were significantly modulated by CDCA. Importantly, CDCA affected distinct microRNAs, including miR-34a, -505, -885, -1260 and -552 that systematically correlated in expression with gene clusters responsible for bile acid, lipid and drug homeostasis incorporating genes, such as e.g. SLCO1B1, SLC22A7, FGF19, CYP2E1, CYP1A2, APO family members and FOXO3. SIGNIFICANCE Bile acids significantly modulate metabolic and drug associated gene networks that are connected to distinct shifts in the microRNAome These findings give novel insights on how BA enfold metabolic and system toxic effects.
Collapse
Affiliation(s)
- Regina Krattinger
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Switzerland
| | - Adrian Boström
- Division of Pharmacology, Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Serene M L Lee
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, Hospital of the University of Munich, Munich, Germany
| | - Wolfgang E Thasler
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, Hospital of the University of Munich, Munich, Germany
| | - Helgi B Schiöth
- Division of Pharmacology, Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Gerd A Kullak-Ublick
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Switzerland.
| | - Jessica Mwinyi
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Switzerland; Division of Pharmacology, Department of Neuroscience, Uppsala University, Uppsala, Sweden
| |
Collapse
|
963
|
Affiliation(s)
- Hooman Allayee
- From the Department of Preventive Medicine (H.A.) and Institute for Genetic Medicine (H.A.), Keck School of Medicine, University of Southern California, Los Angeles, CA; and Departments of Cardiovascular Medicine (S.L.H.) and Cellular and Molecular Medicine, and Center for Cardiovascular Diagnostics and Prevention (S.L.H.), Cleveland Clinic, Cleveland, OH
| | - Stanley L Hazen
- From the Department of Preventive Medicine (H.A.) and Institute for Genetic Medicine (H.A.), Keck School of Medicine, University of Southern California, Los Angeles, CA; and Departments of Cardiovascular Medicine (S.L.H.) and Cellular and Molecular Medicine, and Center for Cardiovascular Diagnostics and Prevention (S.L.H.), Cleveland Clinic, Cleveland, OH.
| |
Collapse
|
964
|
Abdou RM, Zhu L, Baker RD, Baker SS. Gut Microbiota of Nonalcoholic Fatty Liver Disease. Dig Dis Sci 2016; 61:1268-81. [PMID: 26898658 DOI: 10.1007/s10620-016-4045-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 01/16/2016] [Indexed: 02/08/2023]
Abstract
The prevalence of nonalcoholic fatty liver disease has been rapidly increasing worldwide. It has become a leading cause of liver transplantation. Accumulating evidence suggests a significant role for gut microbiota in its development and progression. Here we review the effect of gut microbiota on developing hepatic fatty infiltration and its progression. Current literature supports a possible role for gut microbiota in the development of liver steatosis, inflammation and fibrosis. We also review the literature on possible interventions for NAFLD that target the gut microbiota.
Collapse
Affiliation(s)
- Reham M Abdou
- Digestive Diseases and Nutrition Center, Department of Pediatrics, Women and Children's Hospital of Buffalo, The State University of New York at Buffalo, 219 Bryant Street, Buffalo, NY, 14222, USA.
| | - Lixin Zhu
- Digestive Diseases and Nutrition Center, Department of Pediatrics, Women and Children's Hospital of Buffalo, The State University of New York at Buffalo, 219 Bryant Street, Buffalo, NY, 14222, USA.,, 3435 Main Street, 413 Biomedical Research Building, Buffalo, NY, 14214, USA
| | - Robert D Baker
- Digestive Diseases and Nutrition Center, Department of Pediatrics, Women and Children's Hospital of Buffalo, The State University of New York at Buffalo, 219 Bryant Street, Buffalo, NY, 14222, USA
| | - Susan S Baker
- Digestive Diseases and Nutrition Center, Department of Pediatrics, Women and Children's Hospital of Buffalo, The State University of New York at Buffalo, 219 Bryant Street, Buffalo, NY, 14222, USA
| |
Collapse
|
965
|
Hodge RJ, Nunez DJ. Therapeutic potential of Takeda-G-protein-receptor-5 (TGR5) agonists. Hope or hype? Diabetes Obes Metab 2016; 18:439-43. [PMID: 26818602 DOI: 10.1111/dom.12636] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 12/29/2015] [Accepted: 01/20/2016] [Indexed: 12/27/2022]
Abstract
The gastrointestinal tract regulates glucose and energy metabolism, and there is increasing recognition that bile acids function as key signalling molecules in these processes. For example, bile acid changes that occur after bariatric surgery have been implicated in the effects on satiety, lipid and cholesterol regulation, glucose and energy metabolism, and the gut microbiome. In recent years, Takeda-G-protein-receptor-5 (TGR5), a bile acid receptor found in widely dispersed tissues, has been the target of significant drug discovery efforts in the hope of identifying effective treatments for metabolic diseases including type 2 diabetes, obesity, atherosclerosis, fatty liver disease and cancer. Although the benefits of targeting the TGR5 receptor are potentially great, drug development work to date has identified risks that include histopathological changes, tumorigenesis, gender differences, and questions about the translation of animal data to humans. The present article reviews the noteworthy challenges that must be addressed along the path of development of a safe and effective TGR5 agonist therapy.
Collapse
MESH Headings
- Animals
- Anti-Obesity Agents/adverse effects
- Anti-Obesity Agents/pharmacology
- Anti-Obesity Agents/therapeutic use
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Drug Design
- Drug Discovery/trends
- Drug Evaluation, Preclinical
- Drugs, Investigational/adverse effects
- Drugs, Investigational/pharmacology
- Drugs, Investigational/therapeutic use
- Humans
- Hypoglycemic Agents/adverse effects
- Hypoglycemic Agents/pharmacology
- Hypoglycemic Agents/therapeutic use
- Models, Biological
- Molecular Targeted Therapy
- Obesity/drug therapy
- Obesity/metabolism
- Organ Specificity
- Receptors, G-Protein-Coupled/agonists
- Receptors, G-Protein-Coupled/metabolism
- Translational Research, Biomedical/trends
Collapse
Affiliation(s)
- R J Hodge
- Discovery Medicine, Metabolic Pathways and Cardiovascular Unit, GlaxoSmithKline Research and Development, Research Triangle Park, NC, USA
| | - D J Nunez
- Discovery Medicine, Metabolic Pathways and Cardiovascular Unit, GlaxoSmithKline Research and Development, Research Triangle Park, NC, USA
| |
Collapse
|
966
|
Hegade VS, Speight RA, Etherington RE, Jones DEJ. Novel bile acid therapeutics for the treatment of chronic liver diseases. Therap Adv Gastroenterol 2016; 9:376-91. [PMID: 27134666 PMCID: PMC4830100 DOI: 10.1177/1756283x16630712] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Recent developments in understanding the role of bile acids (BAs) as signalling molecules in human metabolism and inflammation have opened new avenues in the field of hepatology research. BAs are no longer considered as simple molecules helping in fat digestion but as agents with real therapeutic value in treating complex autoimmune and metabolic liver diseases. BAs and their receptors such as farnesoid X receptor, transmembrane G protein-coupled receptor 5 and peroxisome proliferator-activated receptor have been identified as novel targets for drug development. Some of these novel pharmaceuticals are already in clinical evaluation with the most advanced drugs having reached phase III trials. Chronic liver diseases such as primary biliary cholangitis, primary sclerosing cholangitis and nonalcoholic fatty liver disease, for which there is no or limited pharmacotherapy, are most likely to gain from these developments. In this review we discuss recent and the most relevant basic and clinical research findings related to BAs and their implications for novel therapy for chronic liver diseases.
Collapse
Affiliation(s)
| | - R. Alexander Speight
- Institute of Cellular Medicine, Faculty of Medical Science, Newcastle University, Newcastle upon Tyne, UK
| | - Rachel E. Etherington
- Institute of Cellular Medicine, Faculty of Medical Science, Newcastle University, Newcastle upon Tyne, UK
| | - David E. J. Jones
- Institute of Cellular Medicine, Faculty of Medical Science, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
967
|
Mikkelsen KH, Allin KH, Knop FK. Effect of antibiotics on gut microbiota, glucose metabolism and body weight regulation: a review of the literature. Diabetes Obes Metab 2016; 18:444-53. [PMID: 26818734 DOI: 10.1111/dom.12637] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 01/13/2016] [Accepted: 01/20/2016] [Indexed: 12/21/2022]
Abstract
Gut bacteria are involved in a number of host metabolic processes and have been implicated in the development of obesity and type 2 diabetes in humans. The use of antibiotics changes the composition of the gut microbiota and there is accumulating evidence from observational studies for an association between exposure to antibiotics and development of obesity and type 2 diabetes. In the present paper, we review human studies examining the effects of antibiotics on body weight regulation and glucose metabolism and discuss whether the observed findings may relate to alterations in the composition and function of the gut microbiota.
Collapse
Affiliation(s)
- K H Mikkelsen
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - K H Allin
- The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - F K Knop
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
968
|
Kobyliak N, Virchenko O, Falalyeyeva T. Pathophysiological role of host microbiota in the development of obesity. Nutr J 2016; 15:43. [PMID: 27105827 PMCID: PMC4841968 DOI: 10.1186/s12937-016-0166-9] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 04/21/2016] [Indexed: 12/16/2022] Open
Abstract
Overweight and obesity increase the risk for a number of diseases, namely, cardiovascular diseases, type 2 diabetes, dyslipidemia, premature death, non-alcoholic fatty liver disease as well as different types of cancer. Approximately 1.7 billion people in the world suffer from being overweight, most notably in developed countries. Current research efforts have focused on host and environmental factors that may affect energy balance. It was hypothesized that a microbiota profile specific to an obese host with increased energy-yielding behavior may exist. Consequently, the gut microbiota is becoming of significant research interest in relation to obesity in an attempt to better understand the aetiology of obesity and to develop new methods of its prevention and treatment. Alteration of microbiota composition may stimulate development of obesity and other metabolic diseases via several mechanisms: increasing gut permeability with subsequent metabolic inflammation; increasing energy harvest from the diet; impairing short-chain fatty acids synthesis; and altering bile acids metabolism and FXR/TGR5 signaling. Prebiotics and probiotics have physiologic functions that contribute to the health of gut microbiota, maintenance of a healthy body weight and control of factors associated with obesity through their effects on mechanisms that control food intake, body weight, gut microbiota and inflammatory processes.
Collapse
Affiliation(s)
- Nazarii Kobyliak
- Bogomolets National Medical University, T. Shevchenko Boulevard, 13, Kyiv, 01601, Ukraine.
| | - Oleksandr Virchenko
- Taras Shevchenko National University of Kyiv, Volodymyrska Str., 64/13, Kyiv, 01601, Ukraine
| | - Tetyana Falalyeyeva
- Taras Shevchenko National University of Kyiv, Volodymyrska Str., 64/13, Kyiv, 01601, Ukraine
| |
Collapse
|
969
|
Abstract
Glucagon-like peptide-1 (GLP-1) is a peptide hormone, released from intestinal L-cells in response to hormonal, neural and nutrient stimuli. In addition to potentiation of meal-stimulated insulin secretion, GLP-1 signalling exerts numerous pleiotropic effects on various tissues, regulating energy absorption and disposal, as well as cell proliferation and survival. In Type 2 Diabetes (T2D) reduced plasma levels of GLP-1 have been observed, and plasma levels of GLP-1, as well as reduced numbers of GLP-1 producing cells, have been correlated to obesity and insulin resistance. Increasing endogenous secretion of GLP-1 by selective targeting of the molecular mechanisms regulating secretion from the L-cell has been the focus of much recent research. An additional and promising strategy for enhancing endogenous secretion may be to increase the L-cell mass in the intestinal epithelium, but the mechanisms that regulate the growth, survival and function of these cells are largely unknown. We recently showed that prolonged exposure to high concentrations of the fatty acid palmitate induced lipotoxic effects, similar to those operative in insulin-producing cells, in an in vitro model of GLP-1-producing cells. The mechanisms inducing this lipototoxicity involved increased production of reactive oxygen species (ROS). In this review, regulation of GLP-1-secreting cells is discussed, with a focus on the mechanisms underlying GLP-1 secretion, long-term regulation of growth, differentiation and survival under normal as well as diabetic conditions of hypernutrition.
Collapse
|
970
|
Ao M, Domingue JC, Khan N, Javed F, Osmani K, Sarathy J, Rao MC. Lithocholic acid attenuates cAMP-dependent Cl- secretion in human colonic epithelial T84 cells. Am J Physiol Cell Physiol 2016; 310:C1010-23. [PMID: 27076617 DOI: 10.1152/ajpcell.00350.2015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 04/12/2016] [Indexed: 01/14/2023]
Abstract
Bile acids (BAs) play a complex role in colonic fluid secretion. We showed that dihydroxy BAs, but not the monohydroxy BA lithocholic acid (LCA), stimulate Cl(-) secretion in human colonic T84 cells (Ao M, Sarathy J, Domingue J, Alrefai WA, Rao MC. Am J Physiol Cell Physiol 305: C447-C456, 2013). In this study, we explored the effect of LCA on the action of other secretagogues in T84 cells. While LCA (50 μM, 15 min) drastically (>90%) inhibited FSK-stimulated short-circuit current (Isc), it did not alter carbachol-stimulated Isc LCA did not alter basal Isc, transepithelial resistance, cell viability, or cytotoxicity. LCA's inhibitory effect was dose dependent, acted faster from the apical membrane, rapid, and not immediately reversible. LCA also prevented the Isc stimulated by the cAMP-dependent secretagogues 8-bromo-cAMP, lubiprostone, or chenodeoxycholic acid (CDCA). The LCA inhibitory effect was BA specific, since CDCA, cholic acid, or taurodeoxycholic acid did not alter FSK or carbachol action. While LCA alone had no effect on intracellular cAMP concentration ([cAMP]i), it decreased FSK-stimulated [cAMP]i by 90%. Although LCA caused a small increase in intracellular Ca(2+) concentration ([Ca(2+)]i), chelation by BAPTA-AM did not reverse LCA's effect on Isc LCA action does not appear to involve known BA receptors, farnesoid X receptor, vitamin D receptor, muscarinic acetylcholine receptor M3, or bile acid-specific transmembrane G protein-coupled receptor 5. LCA significantly increased ERK1/2 phosphorylation, which was completely abolished by the MEK inhibitor PD-98059. Surprisingly PD-98059 did not reverse LCA's effect on Isc Finally, although LCA had no effect on basal Isc, nystatin permeabilization studies showed that LCA both stimulates an apical cystic fibrosis transmembrane conductance regulator Cl(-) current and inhibits a basolateral K(+) current. In summary, 50 μM LCA greatly inhibits cAMP-stimulated Cl(-) secretion, making low doses of LCA of potential therapeutic interest for diarrheal diseases.
Collapse
Affiliation(s)
- Mei Ao
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois
| | - Jada C Domingue
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois
| | - Nabihah Khan
- Department of Biology, Benedictine University, Lisle, Illinois
| | - Fatima Javed
- Department of Biology, Benedictine University, Lisle, Illinois
| | - Kashif Osmani
- 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 Biology, Benedictine University, Lisle, 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; and
| |
Collapse
|
971
|
|
972
|
Abstract
PURPOSE OF REVIEW This article highlights recent advances in the emerging role that gut microbiota play in modulating metabolic phenotypes, with a particular focus on lipid metabolism. RECENT FINDINGS Accumulating data from both human and animal studies demonstrate that intestinal microbes can affect host lipid metabolism through multiple direct and indirect biological mechanisms. These include a variety of signaling molecules produced by gut bacteria that have potent effects on hepatic lipid and bile metabolism and on reverse cholesterol transport, energy expenditure, and insulin sensitivity in peripheral tissues. Additionally, host genetic factors can modulate the abundance of bacterial taxa, which can subsequently affect various metabolic phenotypes. Proof of causality for identified microbial associations with host lipid-related phenotypes has been demonstrated in several animal studies, but remains a challenge in humans. Ultimately, selective manipulation of the gut microbial ecosystem for intervention will first require a better understanding of which specific bacteria, or alternatively, which bacterial metabolites, are appropriate targets. SUMMARY Recent discoveries have broad implications for elucidating bacterially mediated pathophysiological mechanisms that alter lipid metabolism and other related metabolic traits. From a clinical perspective, this newly recognized endocrine organ system can be targeted for therapeutic benefit of dyslipidemia and cardiometabolic diseases.
Collapse
Affiliation(s)
- Anatole Ghazalpour
- Department of Human Genetics, David Geffen School of Medicine of UCLA, Los Angeles, CA 90095
| | - Ivana Cespedes
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
- Institute for Genetic Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| | - Brian J. Bennett
- Department of Genetics and Nutrition, University of North Carolina, Chapel Hill, NC 27599
- Department of Nutrition Research Institute, University of North Carolina, Chapel Hill, NC 27599
| | - Hooman Allayee
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
- Institute for Genetic Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| |
Collapse
|
973
|
Bolier R, Tolenaars D, Kremer AE, Saris J, Parés A, Verheij J, Bosma PJ, Beuers U, Oude Elferink RP. Enteroendocrine cells are a potential source of serum autotaxin in men. Biochim Biophys Acta Mol Basis Dis 2016; 1862:696-704. [DOI: 10.1016/j.bbadis.2016.01.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 12/22/2015] [Accepted: 01/12/2016] [Indexed: 12/26/2022]
|
974
|
Yan Y, Sha Y, Yao G, Wang S, Kong F, Liu H, Zhang G, Zhang H, Hu C, Zhang X. Roux-en-Y Gastric Bypass Versus Medical Treatment for Type 2 Diabetes Mellitus in Obese Patients: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Medicine (Baltimore) 2016; 95:e3462. [PMID: 27124041 PMCID: PMC4998704 DOI: 10.1097/md.0000000000003462] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The aim of the study is to compare Roux-en-Y gastric bypass (RYGB) surgery versus medical treatment for type 2 diabetes mellitus (T2DM) in obese patients.Bariatric surgery can achieve remission of T2DM in obese patients. RYGB surgery has been performed as one of the most common surgical treatment options for obese patients with T2DM, but the efficacy of RYGB surgery comparing with medical treatment alone has not been conclusively determined.A systematic literature search identified randomized controlled trials (RCTs) evaluating RYGB surgery versus medical treatment for T2DM in obese patients was conducted in PubMed, Embase, Cochrane Database, and Cochrane Clinical Trials Registry. This systematic review and meta-analysis were performed according to the preferred reporting items for systematic reviews and meta-analyses (PRISMA) guidelines. The primary outcome was T2DM remission. Additional analyses comprised hemoglobin A1c (HbA1c), fasting plasma glucose (FPG), body mass index (BMI), waist circumference, serum lipid level, blood pressure, medication use, and adverse events. Random-effects meta-analyses were calculated and presented as weighted odds ratio (OR) or mean difference (MD) with 95% confidence intervals (CI).Six RCTs concerning 410 total obese T2DM patients were included. Follow-up ranged from 12 to 60 months. RYGB surgery was associated with a higher T2DM remission rate (OR: 76.37, 95% CI: 20.70-281.73, P < 0.001) and serum level of high-density lipoprotein cholesterol (MD: 0.24 mmol/L, 95% CI 0.18-0.30 mmol/L, P < 0.001) than medical treatment alone. HbA1c (MD: -1.25%, 95% CI: -1.88% to -0.63%, P < 0.001), BMI (MD: -6.54 kg/m, 95% CI: -9.28 to -3.80 kg/m, P < 0.001), waist circumference (MD: -15.60 cm, 95% CI: -18.21 to -13.00 cm, P < 0.001), triglyceride (MD: -0.87 mmol/L, 95% CI: -1.17 to -0.57 mmol/L, P < 0.001), low-density lipoprotein cholesterol (MD: -0.32 mmol/L, 95% CI: -0.62 to -0.02 mmol/L, P = 0.04), systolic blood pressure (MD: -2.83 mm Hg, 95% CI: -4.88 to -0.78 mm Hg, P < 0.01) were lower after RYGB surgery. However, FPG (MD: -1.58 mmol/L, 95% CI: -3.58 to 0.41 mmol/L, P = 0.12), total cholesterol (MD: -0.40 mmol/L, 95% CI: -0.92 to 0.12 mmol/L, P = 0.13), and diastolic blood pressure (MD: 0.28 mm Hg, 95% CI: -1.89 to 2.45 mm Hg, P = 0.80) were not significantly different between the 2 treatment groups. The medicine use and quality of life were solely improved in the surgical group. Nutritional deficiencies and anemia were noted more frequently in the RYGB group.RYGB surgery is superior to medical treatment for short- to medium-term remission of T2DM, improvement of metabolic condition, and cardiovascular risk factors. Further RCTs should address the safety and long-term benefits of RYGB surgery on obese patients with T2DM.
Collapse
Affiliation(s)
- Yong Yan
- From the Department of General Surgery (YY, GY, SW, FK, HL, GZ, HZ, XZ), Fengxian Central Hospital, Affiliated Hospital of Southern Medical University, Shanghai; Laboratory Medicine Center (YS), Nanfang Hospital, Southern Medical University, Guangzhou; and Department of Endocrinology and Metabolism (CH), Shanghai Diabetes Institute, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
975
|
Malik MY, Jaiswal S, Sharma A, Shukla M, Lal J. Role of enterohepatic recirculation in drug disposition: cooperation and complications. Drug Metab Rev 2016; 48:281-327. [PMID: 26987379 DOI: 10.3109/03602532.2016.1157600] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Enterohepatic recirculation (EHC) concerns many physiological processes and notably affects pharmacokinetic parameters such as plasma half-life and AUC as well as estimates of bioavailability of drugs. Also, EHC plays a detrimental role as the compounds/drugs are allowed to recycle. An in-depth comprehension of this phenomenon and its consequences on the pharmacological effects of affected drugs is important and decisive in the design and development of new candidate drugs. EHC of a compound/drug occurs by biliary excretion and intestinal reabsorption, sometimes with hepatic conjugation and intestinal deconjugation. EHC leads to prolonged elimination half-life of the drugs, altered pharmacokinetics and pharmacodynamics. Study of the EHC of any drug is complicated due to unavailability of the apposite model, sophisticated procedures and ethical concerns. Different in vitro and in vivo methods for studies in experimental animals and humans have been devised, each having its own merits and demerits. Involvement of the different transporters in biliary excretion, intra- and inter-species, pathological and biochemical variabilities obscure the study of the phenomenon. Modeling of drugs undergoing EHC has always been intricate and exigent models have been exploited to interpret the pharmacokinetic profiles of drugs witnessing multiple peaks due to EHC. Here, we critically appraise the mechanisms of bile formation, factors affecting biliary drug elimination, methods to estimate biliary excretion of drugs, EHC, multiple peak phenomenon and its modeling.
Collapse
Affiliation(s)
- Mohd Yaseen Malik
- a Department of Pharmaceutics , National Institute of Pharmaceutical Education and Research (NIPER) , Raebareli , India ;,b Pharmacokinetics & Metabolism Division , CSIR-Central Drug Research Institute , Lucknow , India
| | - Swati Jaiswal
- b Pharmacokinetics & Metabolism Division , CSIR-Central Drug Research Institute , Lucknow , India ;,c Academy of Scientific and Innovative Research , New Delhi , India
| | - Abhisheak Sharma
- b Pharmacokinetics & Metabolism Division , CSIR-Central Drug Research Institute , Lucknow , India ;,c Academy of Scientific and Innovative Research , New Delhi , India ;,d Department of Pharmaceutics and Drug Delivery, School of Pharmacy , The University of Mississippi , Oxford , USA
| | - Mahendra Shukla
- b Pharmacokinetics & Metabolism Division , CSIR-Central Drug Research Institute , Lucknow , India ;,c Academy of Scientific and Innovative Research , New Delhi , India
| | - Jawahar Lal
- b Pharmacokinetics & Metabolism Division , CSIR-Central Drug Research Institute , Lucknow , India ;,c Academy of Scientific and Innovative Research , New Delhi , India
| |
Collapse
|
976
|
Okin D, Medzhitov R. The Effect of Sustained Inflammation on Hepatic Mevalonate Pathway Results in Hyperglycemia. Cell 2016; 165:343-56. [PMID: 26997483 DOI: 10.1016/j.cell.2016.02.023] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 01/12/2016] [Accepted: 02/04/2016] [Indexed: 02/08/2023]
Abstract
Control of plasma glucose level is essential to organismal survival. Sustained inflammation has been implicated in control of glucose homeostasis in cases of infection, obesity, and type 2 diabetes; however, the precise role of inflammation in these complex disease states remains poorly understood. Here, we find that sustained inflammation results in elevated plasma glucose due to increased hepatic glucose production. We find that sustained inflammation suppresses CYP7A1, leading to accumulation of intermediate metabolites at the branch point of the mevalonate pathway. This results in prenylation of RHOC, which is concomitantly induced by inflammatory cytokines. Subsequent activation of RHO-associated protein kinase results in elevated plasma glucose. These findings uncover an unexpected mechanism by which sustained inflammation alters glucose homeostasis.
Collapse
Affiliation(s)
- Daniel Okin
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Ruslan Medzhitov
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
977
|
Brubaker PL, Gil-Lozano M. Glucagon-like peptide-1: The missing link in the metabolic clock? J Diabetes Investig 2016; 7 Suppl 1:70-5. [PMID: 27186359 PMCID: PMC4854508 DOI: 10.1111/jdi.12477] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 12/09/2015] [Accepted: 01/06/2016] [Indexed: 01/10/2023] Open
Abstract
Circadian expression of clock genes in peripheral tissues is critical to the coordinated regulation of intestinal digestive and absorptive functions, insulin secretion, and peripheral tissue nutrient deposition during periods of nutrient ingestion, thereby preventing metabolic dysregulation. As glucagon-like peptide-1 is a key incretin hormone that regulates glucose-dependent insulin secretion, we hypothesized that this intestinal hormone is a player in the peripheral metabolic clock, linking nutrient ingestion to insulin secretion. We have now established that secretion of glucagon-like peptide-1 from the intestinal L cell shows a rhythmic pattern in rats and humans in vivo that is altered by circadian disruptors, such as constant light exposure, consumption of a Western diet and feeding at inappropriate times (i.e., during the light period in rodents). Interestingly, the alterations in the rhythm of the glucagon-like peptide-1 secretory responses were found to parallel the changes in the pattern of insulin responses in association with significant impairments in glucose tolerance. Furthermore, we have detected circadian clock gene expression, and showed circadian secretion of glucagon-like peptide-1 from both the murine and human L cell in vitro. These findings demonstrate that glucagon-like peptide-1 is a functional component of the peripheral metabolic clock, and suggest that altered release of glucagon-like peptide-1 might play a role in the metabolic perturbations that result from circadian disruption.
Collapse
Affiliation(s)
- Patricia L Brubaker
- Department of PhysiologyUniversity of TorontoTorontoOntarioCanada; Department of MedicineUniversity of TorontoTorontoOntarioCanada
| | - Manuel Gil-Lozano
- Department of Physiology University of Toronto Toronto Ontario Canada
| |
Collapse
|
978
|
Yang Z, Zhao A, Li Z, Ge H, Li T, Zhang F, Zhan H, Wang J. Metabolomics reveals positive acceleration(+Gz)-induced metabolic perturbations and the protective effect of Ginkgo biloba extract in a rat model based on ultra high-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry. J Pharm Biomed Anal 2016; 125:77-84. [PMID: 27010354 DOI: 10.1016/j.jpba.2016.03.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 03/02/2016] [Accepted: 03/04/2016] [Indexed: 01/19/2023]
Abstract
Positive acceleration (+Gz) in the head-to-foot direction generated by modern high-performance fighter jets during flight maneuvers is characterized by high G values and a rapid rate of acceleration, and is often long in duration and a repeated occurrence. The acceleration overload far exceeds the pilot's physiological tolerance limits and causes considerable strain on several organ systems. Despite the importance of monitoring pathophysiological alterations related to +Gz exposure, we lack a complete explanation of the pathophysiology of +Gz exposure. Ginkgo biloba extract (GBE) is a classic traditional Chinese medicine (TCM) that might exert a protective effect against +Gz exposure. However, its mechanism remains unclear. Here, a metabolomics approach based on ultra high-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry (UHPLC-Q-TOFMS) was used to characterize +Gz-induced metabolic fluctuations in a rat model and to evaluate the protective effect of GBE. Using partial least-squares discriminant analysis for the classification and selection of biomarkers, eighteen serum metabolites related to +Gz exposure were identified, and were found to primarily involve the fatty acid β-oxidation pathway, glycerophospholipid metabolism, phospholipid metabolism, bile acid metabolism, purine metabolism and lysine metabolism. Taking these potential biomarkers as screening indexes, we found that GBE could reverse the pathological process of +Gz exposure by partially regulating the perturbed fatty acid β-oxidation pathway, glycerophospholipid metabolism, purine metabolism and lysine metabolism. This indicates that UHPLC-Q-TOFMS-based metabolomics provides a powerful tool to reveal serum metabolic fluctuations in response to +Gz exposure and to study the mechanism underlying TCM.
Collapse
Affiliation(s)
- Zhihui Yang
- Department of Clinical Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032 Shanxi, China; Department of Pharmacology, General Hospital of Air Force, PLA, Beijing 100142, China
| | - Andong Zhao
- Department of Clinical Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032 Shanxi, China; Institute of Aviation Medicine, Air Force, Beijing 100142, China
| | - Zhongdong Li
- Department of Pharmacology, General Hospital of Air Force, PLA, Beijing 100142, China
| | - Hua Ge
- Institute of Aviation Medicine, Air Force, Beijing 100142, China
| | - Tonghua Li
- Department of Clinical Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032 Shanxi, China
| | - Fucheng Zhang
- Department of Pharmacology, General Hospital of Air Force, PLA, Beijing 100142, China
| | - Hao Zhan
- Institute of Aviation Medicine, Air Force, Beijing 100142, China.
| | - Jianchang Wang
- Center of Clinical Aviation Medicine, General Hospital of Air Force, PLA, Beijing 100142, China.
| |
Collapse
|
979
|
Abstract
Obesity and insulin resistance are associated with chronic inflammation in metabolic tissues such as adipose tissue and the liver. Recently, growing evidence has implicated the intestinal immune system as an important contributor to metabolic disease. Obesity predisposes to altered intestinal immunity and is associated with changes to the gut microbiota, intestinal barrier function, gut-residing innate and adaptive immune cells, and oral tolerance to luminal antigens. Accordingly, the gut immune system may represent a novel therapeutic target for systemic inflammation in insulin resistance. This review discusses the emerging field of intestinal immunity in obesity-related insulin resistance and how it affects metabolic disease.
Collapse
Affiliation(s)
- Daniel A Winer
- Diabetes Research Group, Division of Cellular and Molecular Biology, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, ON M5G 1L7, Canada; Department of Immunology, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Department of Pathology, University Health Network, 200 Elizabeth Street, Toronto, ON M5G 2C4, Canada.
| | - Helen Luck
- Diabetes Research Group, Division of Cellular and Molecular Biology, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, ON M5G 1L7, Canada; Department of Immunology, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Sue Tsai
- Diabetes Research Group, Division of Cellular and Molecular Biology, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Shawn Winer
- Diabetes Research Group, Division of Cellular and Molecular Biology, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, ON M5G 1L7, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Department of Laboratory Medicine, St. Michael's Hospital, 30 Bond Street, Toronto, ON M5B 1W8, Canada.
| |
Collapse
|
980
|
Gut microbiota-involved mechanisms in enhancing systemic exposure of ginsenosides by coexisting polysaccharides in ginseng decoction. Sci Rep 2016; 6:22474. [PMID: 26932472 PMCID: PMC4774164 DOI: 10.1038/srep22474] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 02/16/2016] [Indexed: 12/21/2022] Open
Abstract
Oral decoctions of traditional Chinese medicines (TCMs) serve for therapeutic and prophylactic management of diseases for centuries. Small molecules and polysaccharides are the dominant chemicals co-occurred in the TCM decoction. Small molecules are well-studied by multidisciplinary elaborations, whereas the role of polysaccharides remains largely elusive. Here we explore a gut microbiota-involved mechanism by which TCM polysaccharides restore the homeostasis of gut microbiota and consequently promote the systemic exposure of concomitant small molecules in the decoction. As a case study, ginseng polysaccharides and ginsenosides in Du-Shen-Tang, the decoction of ginseng, were investigated on an over-fatigue and acute cold stress model. The results indicated that ginseng polysaccharides improved intestinal metabolism and absorption of certain ginsenosides, meanwhile reinstated the perturbed holistic gut microbiota, and particularly enhanced the growth of Lactobacillus spp. and Bacteroides spp., two major metabolic bacteria of ginsenosides. By exploring the synergistic actions of polysaccharides with small molecules, these findings shed new light on scientization and rationalization of the classic TCM decoctions in human health care.
Collapse
|
981
|
Abstract
Bile acids (BA), long believed to only have lipid-digestive functions, have emerged as novel metabolic modulators. They have important endocrine effects through multiple cytoplasmic as well as nuclear receptors in various organs and tissues. BA affect multiple functions to control energy homeostasis, as well as glucose and lipid metabolism, predominantly by activating the nuclear farnesoid X receptor and the cytoplasmic G protein-coupled BA receptor TGR5 in a variety of tissues. However, BA also are aimed at many other cellular targets in a wide array of organs and cell compartments. Their role in the pathogenesis of diabetes, obesity and other 'diseases of civilization' becomes even more clear. They also interact with the gut microbiome, with important clinical implications, further extending the complexity of their biological functions. Therefore, it is not surprising that BA metabolism is substantially modulated by bariatric surgery, a phenomenon contributing favorably to the therapeutic effects of these surgical procedures. Based on these data, several therapeutic approaches to ameliorate obesity and diabetes have been proposed to affect the cellular targets of BA.
Collapse
Affiliation(s)
- Libor Vítek
- Fourth Department of Internal MedicineFirst Faculty of Medicine, Charles University, Na Bojišti 3, Prague 2 12000, Czech RepublicInstitute of Medical Biochemistry and Laboratory DiagnosticsFirst Faculty of Medicine, Charles University, Prague, Czech RepublicInstitute of EndocrinologyCharles University, Prague, Czech Republic Fourth Department of Internal MedicineFirst Faculty of Medicine, Charles University, Na Bojišti 3, Prague 2 12000, Czech RepublicInstitute of Medical Biochemistry and Laboratory DiagnosticsFirst Faculty of Medicine, Charles University, Prague, Czech RepublicInstitute of EndocrinologyCharles University, Prague, Czech Republic
| | - Martin Haluzík
- Fourth Department of Internal MedicineFirst Faculty of Medicine, Charles University, Na Bojišti 3, Prague 2 12000, Czech RepublicInstitute of Medical Biochemistry and Laboratory DiagnosticsFirst Faculty of Medicine, Charles University, Prague, Czech RepublicInstitute of EndocrinologyCharles University, Prague, Czech Republic Fourth Department of Internal MedicineFirst Faculty of Medicine, Charles University, Na Bojišti 3, Prague 2 12000, Czech RepublicInstitute of Medical Biochemistry and Laboratory DiagnosticsFirst Faculty of Medicine, Charles University, Prague, Czech RepublicInstitute of EndocrinologyCharles University, Prague, Czech Republic
| |
Collapse
|
982
|
Abstract
Metformin is an effective agent with a good safety profile that is widely used as a first-line treatment for type 2 diabetes, yet its mechanisms of action and variability in terms of efficacy and side effects remain poorly understood. Although the liver is recognised as a major site of metformin pharmacodynamics, recent evidence also implicates the gut as an important site of action. Metformin has a number of actions within the gut. It increases intestinal glucose uptake and lactate production, increases GLP-1 concentrations and the bile acid pool within the intestine, and alters the microbiome. A novel delayed-release preparation of metformin has recently been shown to improve glycaemic control to a similar extent to immediate-release metformin, but with less systemic exposure. We believe that metformin response and tolerance is intrinsically linked with the gut. This review examines the passage of metformin through the gut, and how this can affect the efficacy of metformin treatment in the individual, and contribute to the side effects associated with metformin intolerance.
Collapse
Affiliation(s)
- Laura J McCreight
- Pearson Group, Division of Cardiovascular and Diabetes Medicine, School of Medicine, University of Dundee, Ninewells Hospital, Mailbox 12, Level 5, Dundee, DD1 9SY, UK
| | - Clifford J Bailey
- School of Life and Health Sciences, Aston University, Birmingham, UK
| | - Ewan R Pearson
- Pearson Group, Division of Cardiovascular and Diabetes Medicine, School of Medicine, University of Dundee, Ninewells Hospital, Mailbox 12, Level 5, Dundee, DD1 9SY, UK.
| |
Collapse
|
983
|
Morimoto K, Watanabe M, Sugizaki T, Irie JI, Itoh H. Intestinal Bile Acid Composition Modulates Prohormone Convertase 1/3 (PC1/3) Expression and Consequent GLP-1 Production in Male Mice. Endocrinology 2016; 157:1071-81. [PMID: 26789236 DOI: 10.1210/en.2015-1551] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Besides an established medication for hypercholesterolemia, bile acid binding resins (BABRs) present antidiabetic effects. Although the mechanisms underlying these effects are still enigmatic, glucagon-like peptide-1 (GLP-1) appears to be involved. In addition to a few reported mechanisms, we propose prohormone convertase 1/3 (PC1/3), an essential enzyme of GLP-1 production, as a potent molecule in the GLP-1 release induced by BABRs. In our study, the BABR colestimide leads to a bile acid-specific G protein-coupled receptor TGR5-dependent induction of PC1/3 gene expression. Here, we focused on the alteration of intestinal bile acid composition and consequent increase of total TGR5 agonistic activity to explain the TGR5 activation. Furthermore, we demonstrate that nuclear factor of activated T cells mediates the TGR5-triggered PC1/3 gene expression. Altogether, our data indicate that the TGR5-dependent intestinal PC1/3 gene expression supports the BABR-stimulated GLP-1 release. We also propose a combination of BABR and dipeptidyl peptidase-4 inhibitor in the context of GLP-1-based antidiabetic therapy.
Collapse
Affiliation(s)
- Kohkichi Morimoto
- Department of Internal Medicine (K.M., T.S., J.-i.I., H.I.), School of Medicine, Keio University, Tokyo 160-8582, Japan; and Graduate School of Media and Governance (M.W.), Faculty of Environment and Information Studies, Keio University, Kanagawa 252-0882, Japan
| | - Mitsuhiro Watanabe
- Department of Internal Medicine (K.M., T.S., J.-i.I., H.I.), School of Medicine, Keio University, Tokyo 160-8582, Japan; and Graduate School of Media and Governance (M.W.), Faculty of Environment and Information Studies, Keio University, Kanagawa 252-0882, Japan
| | - Taichi Sugizaki
- Department of Internal Medicine (K.M., T.S., J.-i.I., H.I.), School of Medicine, Keio University, Tokyo 160-8582, Japan; and Graduate School of Media and Governance (M.W.), Faculty of Environment and Information Studies, Keio University, Kanagawa 252-0882, Japan
| | - Jun-ichiro Irie
- Department of Internal Medicine (K.M., T.S., J.-i.I., H.I.), School of Medicine, Keio University, Tokyo 160-8582, Japan; and Graduate School of Media and Governance (M.W.), Faculty of Environment and Information Studies, Keio University, Kanagawa 252-0882, Japan
| | - Hiroshi Itoh
- Department of Internal Medicine (K.M., T.S., J.-i.I., H.I.), School of Medicine, Keio University, Tokyo 160-8582, Japan; and Graduate School of Media and Governance (M.W.), Faculty of Environment and Information Studies, Keio University, Kanagawa 252-0882, Japan
| |
Collapse
|
984
|
Sun W, Zhang D, Wang Z, Sun J, Xu B, Chen Y, Ding L, Huang X, Lv X, Lu J, Bi Y, Xu Q. Insulin Resistance is Associated With Total Bile Acid Level in Type 2 Diabetic and Nondiabetic Population: A Cross-Sectional Study. Medicine (Baltimore) 2016; 95:e2778. [PMID: 26962776 PMCID: PMC4998857 DOI: 10.1097/md.0000000000002778] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 12/31/2015] [Accepted: 01/19/2016] [Indexed: 02/06/2023] Open
Abstract
Bile acid metabolism was reported to be involved in glucose metabolism homeostasis. However, the exact relationship between bile acid and glucose metabolism as well as insulin sensitivity is not clarified. Therefore, we sought to investigate the association between insulin sensitivity and hyperbileacidemia in type 2 diabetic and nondiabetic population.This community-based cross-sectional study included 9603 residents from Jiading, Shanghai, China, who were 40 years and older. Standardized questionnaire, anthropometric measurements and laboratory tests were conducted. Homeostasis model assessment of insulin resistance (HOMA-IR) ≥ 2.7 was defined as insulin resistance and fasting TBA ≥ 10 mmol/L was defined as hyperbileacidemia.Multivariate stepwise regression analysis revealed that HOMA-IR, age, and male sex were positively associated with hyperbileacidemia in both nondiabetic and diabetic participants. In multivariate logistic models, participants with insulin resistance had significantly higher risk of hyperbileacidemia compared to those who have no insulin resistance, in both nondiabetic and diabetic population (nondiabetic: OR = 1.76; 95% CI 1.42-2.19; P < 0.001; diabetic: OR = 1.56; 95% CI 1.06 - 2.31; P = 0.025, respectively). Further adjustment for the HbA1c level in diabetic population did not change the significant association (OR = 1.59; 95% CI 1.06 - 2.40; P = 0.024). In nondiabetic participants, each 1-unit increment of HOMA-IR conferred an 18% higher risk of hyperbileacidemia (95% CI 1.04-1.35; P = 0.013), whereas in diabetic participants, this association was similar but not significant (95% CI 0.95-1.59; P = 0.117).Insulin resistance was positively associated with hyperbileacidemia in both nondiabetic and diabetic population. The increase in the bile acid level in insulin-resistant population regardless of status of diabetes and glucose level indicated the important role of insulin resistance in the regulation of bile acid metabolism in human.
Collapse
Affiliation(s)
- Wanwan Sun
- From the National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine (WS, DZ, ZW, JS, BX, YC, LD, XH, XL, JL, YB); Institute of Health Sciences, Shanghai Institutes for Biological Sciences (JS); and Department of Research and Development, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine (QX), Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
985
|
Xu Y. Recent Progress on Bile Acid Receptor Modulators for Treatment of Metabolic Diseases. J Med Chem 2016; 59:6553-79. [DOI: 10.1021/acs.jmedchem.5b00342] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Yanping Xu
- Lilly Research
Laboratories, Eli Lilly and Company, Lilly Corporate Center, DC 1910, Indianapolis, Indiana 46285, United States
| |
Collapse
|
986
|
Bradford EM, Vairamani K, Shull GE. Differential expression of pancreatic protein and chemosensing receptor mRNAs in NKCC1-null intestine. World J Gastrointest Pathophysiol 2016; 7:138-149. [PMID: 26909237 PMCID: PMC4753180 DOI: 10.4291/wjgp.v7.i1.138] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 10/10/2015] [Accepted: 12/18/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the intestinal functions of the NKCC1 Na+-K+-2Cl cotransporter (SLC12a2 gene), differential mRNA expression changes in NKCC1-null intestine were analyzed.
METHODS: Microarray analysis of mRNA from intestines of adult wild-type mice and gene-targeted NKCC1-null mice (n = 6 of each genotype) was performed to identify patterns of differential gene expression changes. Differential expression patterns were further examined by Gene Ontology analysis using the online Gorilla program, and expression changes of selected genes were verified using northern blot analysis and quantitative real time-polymerase chain reaction. Histological staining and immunofluorescence were performed to identify cell types in which upregulated pancreatic digestive enzymes were expressed.
RESULTS: Genes typically associated with pancreatic function were upregulated. These included lipase, amylase, elastase, and serine proteases indicative of pancreatic exocrine function, as well as insulin and regenerating islet genes, representative of endocrine function. Northern blot analysis and immunohistochemistry showed that differential expression of exocrine pancreas mRNAs was specific to the duodenum and localized to a subset of goblet cells. In addition, a major pattern of changes involving differential expression of olfactory receptors that function in chemical sensing, as well as other chemosensing G-protein coupled receptors, was observed. These changes in chemosensory receptor expression may be related to the failure of intestinal function and dependency on parenteral nutrition observed in humans with SLC12a2 mutations.
CONCLUSION: The results suggest that loss of NKCC1 affects not only secretion, but also goblet cell function and chemosensing of intestinal contents via G-protein coupled chemosensory receptors.
Collapse
|
987
|
Eelderink C, Noort MWJ, Sozer N, Koehorst M, Holst JJ, Deacon CF, Rehfeld JF, Poutanen K, Vonk RJ, Oudhuis L, Priebe MG. Difference in postprandial GLP-1 response despite similar glucose kinetics after consumption of wheat breads with different particle size in healthy men. Eur J Nutr 2016; 56:1063-1076. [PMID: 26857762 PMCID: PMC5346412 DOI: 10.1007/s00394-016-1156-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 01/10/2016] [Indexed: 01/08/2023]
Abstract
Purpose Underlying mechanisms of the beneficial health effects of low glycemic index starchy foods are not fully elucidated yet. We varied the wheat particle size to obtain fiber-rich breads with a high and low glycemic response and investigated the differences in postprandial glucose kinetics and metabolic response after their consumption. Methods Ten healthy male volunteers participated in a randomized, crossover study, consuming 13C-enriched breads with different structures; a control bread (CB) made from wheat flour combined with wheat bran, and a kernel bread (KB) where 85 % of flour was substituted with broken wheat kernels. The structure of the breads was characterized extensively. The use of stable isotopes enabled calculation of glucose kinetics: rate of appearance of exogenous glucose, endogenous glucose production, and glucose clearance rate. Additionally, postprandial plasma concentrations of glucose, insulin, glucagon, incretins, cholecystokinin, and bile acids were analyzed. Results Despite the attempt to obtain a bread with a low glycemic response by replacing flour by broken kernels, the glycemic response and glucose kinetics were quite similar after consumption of CB and KB. Interestingly, the glucagon-like peptide-1 (GLP-1) response was much lower after KB compared to CB (iAUC, P < 0.005). A clear postprandial increase in plasma conjugated bile acids was observed after both meals. Conclusions Substitution of 85 % wheat flour by broken kernels in bread did not result in a difference in glucose response and kinetics, but in a pronounced difference in GLP-1 response. Thus, changing the processing conditions of wheat for baking bread can influence the metabolic response beyond glycemia and may therefore influence health. Electronic supplementary material The online version of this article (doi:10.1007/s00394-016-1156-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Coby Eelderink
- Center for Medical Biomics, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands. .,Top Institute Food and Nutrition, Wageningen, The Netherlands.
| | | | - Nesli Sozer
- VTT Technical Research Centre of Finland, Espoo, Finland
| | - Martijn Koehorst
- Department of Laboratory Medicine, Center for Liver Digestive and Metabolic Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jens J Holst
- NNF Center for Basic Metabolic Research, Department of Biomedical Sciences, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Carolyn F Deacon
- NNF Center for Basic Metabolic Research, Department of Biomedical Sciences, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Jens F Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Kaisa Poutanen
- VTT Technical Research Centre of Finland, Espoo, Finland
| | - Roel J Vonk
- Center for Medical Biomics, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Lizette Oudhuis
- Top Institute Food and Nutrition, Wageningen, The Netherlands.,TNO Food and Nutrition, Zeist, The Netherlands
| | - Marion G Priebe
- Center for Medical Biomics, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.,Top Institute Food and Nutrition, Wageningen, The Netherlands
| |
Collapse
|
988
|
Abstract
The incretin hormones glucose-dependent insulinotropic polypeptide (GIP) and glucagon like peptide-1 (GLP-1) are secreted from enteroendocrine cells in the gut and regulate physiological and homeostatic functions related to glucose control, metabolism and food intake. This review provides a systematic summary of the molecular mechanisms underlying secretion from incretin cells, and an understanding of how they sense and interact with lumen and vascular factors and the enteric nervous system through transporters and G-protein coupled receptors (GPCRs) present on their surface to ultimately culminate in hormone release. Some of the molecules described below such as sodium coupled glucose transporter 1 (SGLT1), G-protein coupled receptor (GPR) 119 and GPR40 are targets of novel therapeutics designed to enhance endogenous gut hormone release. Synthetic ligands at these receptors aimed at treating obesity and type 2 diabetes are currently under investigation.
Collapse
Affiliation(s)
- Ramona Pais
- The Wellcome Trust–MRC Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Fiona M. Gribble
- The Wellcome Trust–MRC Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Addenbrookes’s Hospital, Box 289, Hills Road, Cambridge, CB2 0QQ, UK
| | | |
Collapse
|
989
|
SAR studies on FXR modulators led to the discovery of the first combined FXR antagonistic/TGR5 agonistic compound. Future Med Chem 2016; 8:133-48. [PMID: 26824277 DOI: 10.4155/fmc.15.178] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Bile acids can serve as signaling molecules by activating the nuclear receptor FXR and the G-protein-coupled receptor TGR5 and both bile acid receptors are prominent experimental drug targets. Results/methodology: In this study we optimized the fatty acid mimetic compound pirinixic acid to a new scaffold with the aim to develop novel FXR modulatory compounds. After a multistep structure-activity optimization process, we discovered FXR agonistic compounds and the first dual FXR antagonistic and TGR5 agonistic compound 79a. CONCLUSION With this novel dual activity profile on both bile acid receptors 79a might be a valuable pharmalogical tool to further study the bile acid signaling network.
Collapse
|
990
|
Agarwal S, Patil A, Aware U, Deshmukh P, Darji B, Sasane S, Sairam KVV, Priyadarsiny P, Giri P, Patel H, Giri S, Jain M, Desai RC. Discovery of a Potent and Orally Efficacious TGR5 Receptor Agonist. ACS Med Chem Lett 2016; 7:51-5. [PMID: 26819665 PMCID: PMC4716599 DOI: 10.1021/acsmedchemlett.5b00323] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 11/20/2015] [Indexed: 12/13/2022] Open
Abstract
TGR5 is a G protein-coupled receptor (GPCR), activation of which promotes secretion of glucagon-like peptide-1 (GLP-1) and modulates insulin secretion. The 2-thio-imidazole derivative 6g was identified as a novel, potent, and selective TGR5 agonist (hTGR5 EC50 = 57 pM, mTGR5 = 62 pM) with a favorable pharmacokinetic profile. The compound 6g was found to have potent glucose lowering effects in vivo during an oral glucose tolerance test in DIO C57 mice with ED50 of 7.9 mg/kg and ED90 of 29.2 mg/kg.
Collapse
Affiliation(s)
- Sameer Agarwal
- Zydus Research Centre, Cadila
Healthcare Ltd., Sarkhej-Bavla
N.H. No. 8 A, Moraiya, Ahmedabad 382 210, India
| | - Amit Patil
- Zydus Research Centre, Cadila
Healthcare Ltd., Sarkhej-Bavla
N.H. No. 8 A, Moraiya, Ahmedabad 382 210, India
| | - Umesh Aware
- Zydus Research Centre, Cadila
Healthcare Ltd., Sarkhej-Bavla
N.H. No. 8 A, Moraiya, Ahmedabad 382 210, India
| | - Prashant Deshmukh
- Zydus Research Centre, Cadila
Healthcare Ltd., Sarkhej-Bavla
N.H. No. 8 A, Moraiya, Ahmedabad 382 210, India
| | - Brijesh Darji
- Zydus Research Centre, Cadila
Healthcare Ltd., Sarkhej-Bavla
N.H. No. 8 A, Moraiya, Ahmedabad 382 210, India
| | - Santosh Sasane
- Zydus Research Centre, Cadila
Healthcare Ltd., Sarkhej-Bavla
N.H. No. 8 A, Moraiya, Ahmedabad 382 210, India
| | - Kalapatapu V. V.
M. Sairam
- Zydus Research Centre, Cadila
Healthcare Ltd., Sarkhej-Bavla
N.H. No. 8 A, Moraiya, Ahmedabad 382 210, India
| | - Priyanka Priyadarsiny
- Zydus Research Centre, Cadila
Healthcare Ltd., Sarkhej-Bavla
N.H. No. 8 A, Moraiya, Ahmedabad 382 210, India
| | - Poonam Giri
- Zydus Research Centre, Cadila
Healthcare Ltd., Sarkhej-Bavla
N.H. No. 8 A, Moraiya, Ahmedabad 382 210, India
| | - Harilal Patel
- Zydus Research Centre, Cadila
Healthcare Ltd., Sarkhej-Bavla
N.H. No. 8 A, Moraiya, Ahmedabad 382 210, India
| | - Suresh Giri
- Zydus Research Centre, Cadila
Healthcare Ltd., Sarkhej-Bavla
N.H. No. 8 A, Moraiya, Ahmedabad 382 210, India
| | - Mukul Jain
- Zydus Research Centre, Cadila
Healthcare Ltd., Sarkhej-Bavla
N.H. No. 8 A, Moraiya, Ahmedabad 382 210, India
| | - Ranjit C. Desai
- Zydus Research Centre, Cadila
Healthcare Ltd., Sarkhej-Bavla
N.H. No. 8 A, Moraiya, Ahmedabad 382 210, India
| |
Collapse
|
991
|
Francino MP. Antibiotics and the Human Gut Microbiome: Dysbioses and Accumulation of Resistances. Front Microbiol 2016; 6:1543. [PMID: 26793178 PMCID: PMC4709861 DOI: 10.3389/fmicb.2015.01543] [Citation(s) in RCA: 506] [Impact Index Per Article: 56.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 12/21/2015] [Indexed: 12/12/2022] Open
Abstract
The human microbiome is overly exposed to antibiotics, due, not only to their medical use, but also to their utilization in farm animals and crops. Microbiome composition can be rapidly altered by exposure to antibiotics, with potential immediate effects on health, for instance through the selection of resistant opportunistic pathogens that can cause acute disease. Microbiome alterations induced by antibiotics can also indirectly affect health in the long-term. The mutualistic microbes in the human body interact with many physiological processes, and participate in the regulation of immune and metabolic homeostasis. Therefore, antibiotic exposure can alter many basic physiological equilibria, promoting long-term disease. In addition, excessive antibiotic use fosters bacterial resistance, and the overly exposed human microbiome has become a significant reservoir of resistance genes, contributing to the increasing difficulty in controlling bacterial infections. Here, the complex relationships between antibiotics and the human microbiome are reviewed, with focus on the intestinal microbiota, addressing (1) the effects of antibiotic use on the composition and function of the gut microbiota, (2) the impact of antibiotic-induced microbiota alterations on immunity, metabolism, and health, and (3) the role of the gut microbiota as a reservoir of antibiotic resistances.
Collapse
Affiliation(s)
- M P Francino
- Unitat Mixta d'Investigació en Genòmica i Salut, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO)-Salud Pública/Institut Cavanilles de Biodiversitat i Biologia Evolutiva, Universitat de ValènciaValència, Spain; Consorcio de Investigación Biomédica en Red de Epidemiología y Salud PúblicaMadrid, Spain
| |
Collapse
|
992
|
Corcelles R, Daigle CR, Schauer PR. MANAGEMENT OF ENDOCRINE DISEASE: Metabolic effects of bariatric surgery. Eur J Endocrinol 2016; 174:R19-28. [PMID: 26340972 DOI: 10.1530/eje-15-0533] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 09/03/2015] [Indexed: 12/13/2022]
Abstract
Obesity is associated with an increased risk of type 2 diabetes, hypertension, dyslipidemia, cardiovascular disease, osteoarthritis, numerous cancers and increased mortality. It is estimated that at least 2.8 million adults die each year due to obesity-related cardiovascular disease. Increasing in parallel with the global obesity problem is metabolic syndrome, which has also reached epidemic levels. Numerous studies have demonstrated that bariatric surgery is associated with significant and durable weight loss with associated improvement of obesity-related comorbidities. This review aims to summarize the effects of bariatric surgery on the components of metabolic syndrome (hyperglycemia, hyperlipidemia and hypertension), weight loss, perioperative morbidity and mortality, and the long-term impact on cardiovascular risk and mortality.
Collapse
Affiliation(s)
- Ricard Corcelles
- Bariatric and Metabolic InstituteCleveland Clinic, 9500 Euclid Avenue, M61 Cleveland, OH 44195, USADepartment of Gastrointestinal SurgeryInstitute of Digestive and Metabolic Diseases, Fundació Clínic per a la Recerca Biomèdica, Hospital Clinic de Barcelona, Barcelona, Spain Bariatric and Metabolic InstituteCleveland Clinic, 9500 Euclid Avenue, M61 Cleveland, OH 44195, USADepartment of Gastrointestinal SurgeryInstitute of Digestive and Metabolic Diseases, Fundació Clínic per a la Recerca Biomèdica, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Christopher R Daigle
- Bariatric and Metabolic InstituteCleveland Clinic, 9500 Euclid Avenue, M61 Cleveland, OH 44195, USADepartment of Gastrointestinal SurgeryInstitute of Digestive and Metabolic Diseases, Fundació Clínic per a la Recerca Biomèdica, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Philip R Schauer
- Bariatric and Metabolic InstituteCleveland Clinic, 9500 Euclid Avenue, M61 Cleveland, OH 44195, USADepartment of Gastrointestinal SurgeryInstitute of Digestive and Metabolic Diseases, Fundació Clínic per a la Recerca Biomèdica, Hospital Clinic de Barcelona, Barcelona, Spain
| |
Collapse
|
993
|
Abstract
The armamentarium for the treatment of dyslipidemia today comprises six different modes of action with overall around 24 different drugs. The treatment of lipid disorders was revolutionized with the introduction of statins which have become the most important therapeutic option available today to reduce and prevent atherosclerosis and its detrimental consequences like cardiovascular diseases and stroke. With and optimized reduction of elevated LDL levels with statins, the risk for cardiovascular diseases (CVD) can be reduced by 30%, indicating a residual remaining risk of 70% for the development and progression of CVD notifying still a high medical need for more effective antilipidemic drugs. Consequently, the search for novel lipid-modifying drugs is still one of the most active areas in research and development in the pharmaceutical industry. Major focus lies on approaches to LDL-lowering drugs superior to statins with regard to efficacy, safety, and patient compliance and on approaches modifying plasma levels and functionality of HDL particles based on the clinically validated inverse relationship between high-plasma HDL levels and the risk for CVD. The available drugs today for the treatment of dyslipidemia are small organic molecules or nonabsorbable polymers for binding of bile acids to be applied orally. Besides small molecules for novel targets, biological drugs such as monoclonal antibodies, antisense or gene-silencing oligonucleotides, peptidomimetics, reconstituted synthetic HDL particles and therapeutic proteins are novel approaches in clinical development are which have to be applied by injection or infusion. The promising clinical results of several novel drug candidates, particularly for LDL cholesterol lowering with monoclonal antibodies raised against PCSK9, may indicate more than a decade after the statins, the entrance of new breakthrough therapies to treat lipid disorders.
Collapse
Affiliation(s)
- Werner Kramer
- Institute of Biochemistry, Biocenter, Goethe-Universität Frankfurt, Max-von-Laue-Str. 9, Frankfurt, Germany.
| |
Collapse
|
994
|
Khan MJ, Gerasimidis K, Edwards CA, Shaikh MG. Role of Gut Microbiota in the Aetiology of Obesity: Proposed Mechanisms and Review of the Literature. J Obes 2016; 2016:7353642. [PMID: 27703805 PMCID: PMC5040794 DOI: 10.1155/2016/7353642] [Citation(s) in RCA: 172] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 05/21/2016] [Accepted: 08/21/2016] [Indexed: 02/06/2023] Open
Abstract
The aetiology of obesity has been attributed to several factors (environmental, dietary, lifestyle, host, and genetic factors); however none of these fully explain the increase in the prevalence of obesity worldwide. Gut microbiota located at the interface of host and environment in the gut are a new area of research being explored to explain the excess accumulation of energy in obese individuals and may be a potential target for therapeutic manipulation to reduce host energy storage. Several mechanisms have been suggested to explain the role of gut microbiota in the aetiology of obesity such as short chain fatty acid production, stimulation of hormones, chronic low-grade inflammation, lipoprotein and bile acid metabolism, and increased endocannabinoid receptor system tone. However, evidence from animal and human studies clearly indicates controversies in determining the cause or effect relationship between the gut microbiota and obesity. Metagenomics based studies indicate that functionality rather than the composition of gut microbiota may be important. Further mechanistic studies controlling for environmental and epigenetic factors are therefore required to help unravel obesity pathogenesis.
Collapse
Affiliation(s)
- Muhammad Jaffar Khan
- Institute of Basic Medical Sciences, Khyber Medical University, Phase V Hayatabad, Peshawar, Khyber Pakhtunkhwa, Pakistan
- Human Nutrition, School of Medicine, Dentistry and Nursing, College of Medical Veterinary and Life Sciences, University of Glasgow, Level 3, New Lister Building, Glasgow Royal Infirmary, 10-16 Alexandra Parade, Glasgow G31 2ER, UK
- *Muhammad Jaffar Khan:
| | - Konstantinos Gerasimidis
- Human Nutrition, School of Medicine, Dentistry and Nursing, College of Medical Veterinary and Life Sciences, University of Glasgow, Level 3, New Lister Building, Glasgow Royal Infirmary, 10-16 Alexandra Parade, Glasgow G31 2ER, UK
| | - Christine Ann Edwards
- Human Nutrition, School of Medicine, Dentistry and Nursing, College of Medical Veterinary and Life Sciences, University of Glasgow, Level 3, New Lister Building, Glasgow Royal Infirmary, 10-16 Alexandra Parade, Glasgow G31 2ER, UK
| | - M. Guftar Shaikh
- Department of Endocrinology, Royal Hospital for Children, 1345 Govan Rd, Govan, Glasgow G51 4TF, UK
| |
Collapse
|
995
|
Singh V, Yeoh BS, Ntambi JM, Vijay-Kumar M. Influence of Gut Microbiota on Hepatic Lipogenesis and Disease Pathogenesis. HEPATIC DE NOVO LIPOGENESIS AND REGULATION OF METABOLISM 2016:189-209. [DOI: 10.1007/978-3-319-25065-6_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
996
|
Abstract
Metabolomics is a promising approach for the identification of chemical compounds that serve for early detection, diagnosis, prediction of therapeutic response and prognosis of disease. Moreover, metabolomics has shown to increase the diagnostic threshold and prediction of type 2 diabetes. Evidence suggests that branched-chain amino acids, acylcarnitines and aromatic amino acids may play an early role on insulin resistance, exposing defects on amino acid metabolism, β-oxidation, and tricarboxylic acid cycle. This review aims to provide a panoramic view of the metabolic shifts that antecede or follow type 2 diabetes. Key messages BCAAs, AAAs and acylcarnitines are strongly associated with early insulin resistance. Diabetes risk prediction has been improved when adding metabolomic markers of dysglycemia to standard clinical and biochemical factors.
Collapse
Affiliation(s)
| | - Carlos A Aguilar-Salinas
- a Instituto Nacional De Ciencias Médicas Y Nutrición "Salvador Zubirán" , Ciudad De México , D.F
| | - Ivette Cruz-Bautista
- a Instituto Nacional De Ciencias Médicas Y Nutrición "Salvador Zubirán" , Ciudad De México , D.F
| | | |
Collapse
|
997
|
Keitel V, Reich M, Häussinger D. TGR5: pathogenetic role and/or therapeutic target in fibrosing cholangitis? Clin Rev Allergy Immunol 2016; 48:218-25. [PMID: 25138774 DOI: 10.1007/s12016-014-8443-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Primary sclerosing cholangitis (PSC) is a chronic inflammatory disease affecting the intrahepatic and extrahepatic biliary tree leading to bile duct strictures, progressive cholestasis, and development of liver fibrosis and cirrhosis. The pathogenesis of PSC is still elusive; however, both an immune-mediated injury of the bile ducts as well as increased recruitment of intestinal-primed T lymphocytes to the biliary tracts seem to contribute to disease development and progression. TGR5 (Gpbar-1) is a G-protein-coupled receptor responsive to bile acids, which is expressed in cholangiocytes, intestinal epithelial cells, and macrophages of the liver and intestine as well as in CD14-positive monocytes of the peripheral blood. Activation of TGR5 in biliary epithelial cells promotes chloride and bicarbonate secretion, triggers cell proliferation, and prevents apoptotic cell death. In immune cells, stimulation of TGR5 inhibits cytokine expression and secretion, thus reducing systemic as well as hepatic and intestinal inflammation. The expression pattern of TGR5 in the liver and intestine as well as the potential protective functions of TGR5 suggest a role for this receptor in the pathogenesis of PSC. While mutations in the coding region of the TGR5 gene are too rare to contribute to overall disease susceptibility, the expression and localization of the receptor have not been studied in PSC livers. Pharmacological activation of TGR5 in mice promotes protective mechanisms in biliary epithelial cells and reduces hepatic and systemic inflammation; however, it also provokes pruritus. Further studies are needed to predict the potential benefits as well as side effects of TGR5 agonist treatment in PSC patients.
Collapse
Affiliation(s)
- Verena Keitel
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany,
| | | | | |
Collapse
|
998
|
Duodenal-Jejunal Bypass Preferentially Elevates Serum Taurine-Conjugated Bile Acids and Alters Gut Microbiota in a Diabetic Rat Model. Obes Surg 2015; 26:1890-9. [DOI: 10.1007/s11695-015-2031-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
999
|
Pharmacology of bile acid receptors: Evolution of bile acids from simple detergents to complex signaling molecules. Pharmacol Res 2015; 104:9-21. [PMID: 26706784 DOI: 10.1016/j.phrs.2015.12.007] [Citation(s) in RCA: 175] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Accepted: 12/03/2015] [Indexed: 12/17/2022]
Abstract
For many years, bile acids were thought to only function as detergents which solubilize fats and facilitate the uptake of fat-soluble vitamins in the intestine. Many early observations; however, demonstrated that bile acids regulate more complex processes, such as bile acids synthesis and immune cell function through activation of signal transduction pathways. These studies were the first to suggest that receptors may exist for bile acids. Ultimately, seminal studies by many investigators led to the discovery of several bile acid-activated receptors including the farnesoid X receptor, the vitamin D receptor, the pregnane X receptor, TGR5, α5 β1 integrin, and sphingosine-1-phosphate receptor 2. Several of these receptors are expressed outside of the gastrointestinal system, indicating that bile acids may have diverse functions throughout the body. Characterization of the functions of these receptors over the last two decades has identified many important roles for these receptors in regulation of bile acid synthesis, transport, and detoxification; regulation of glucose utilization; regulation of fatty acid synthesis and oxidation; regulation of immune cell function; regulation of energy expenditure; and regulation of neural processes such as gastric motility. Through these many functions, bile acids regulate many aspects of digestion ranging from uptake of essential vitamins to proper utilization of nutrients. Accordingly, within a short time period, bile acids moved beyond simple detergents and into the realm of complex signaling molecules. Because of the important processes that bile acids regulate through activation of receptors, drugs that target these receptors are under development for the treatment of several diseases, including cholestatic liver disease and metabolic syndrome. In this review, we will describe the various bile acid receptors, the signal transduction pathways activated by these receptors, and briefly discuss the physiological processes that these receptors regulate.
Collapse
|
1000
|
Park BO, Kim SH, Kong GY, Kim DH, Kwon MS, Lee SU, Kim MO, Cho S, Lee S, Lee HJ, Han SB, Kwak YS, Lee SB, Kim S. Selective novel inverse agonists for human GPR43 augment GLP-1 secretion. Eur J Pharmacol 2015; 771:1-9. [PMID: 26683635 DOI: 10.1016/j.ejphar.2015.12.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 11/17/2015] [Accepted: 12/07/2015] [Indexed: 12/25/2022]
Abstract
GPR43/Free Fatty Acid Receptor 2 (FFAR2) is known to be activated by short-chain fatty acids and be coupled to Gi and Gq family of heterotrimeric G proteins. GPR43 is mainly expressed in neutrophils, adipocytes and enteroendocrine cells, implicated to be involved in inflammation, obesity and type 2 diabetes. However, several groups have reported the contradictory data about the physiological functions of GPR43, so that its roles in vivo remain unclear. Here, we demonstrate that a novel compound of pyrimidinecarboxamide class named as BTI-A-404 is a selective and potent competitive inverse agonist of human GPR43, but not the murine ortholog. Through structure-activity relationship (SAR), we also found active compound named as BTI-A-292. These regulators increased the cyclic AMP level and reduced acetate-induced cytoplasmic Ca(2+) level. Furthermore, we show that they modulated the downstream signaling pathways of GPR43, such as ERK, p38 MAPK, and NF-κB. It was surprising that two compounds augmented the secretion of glucagon-like peptide 1 (GLP-1) in NCI-H716 cell line. Collectively, these novel and specific competitive inhibitors regulate all aspects of GPR43 signaling and the results underscore the therapeutic potential of them.
Collapse
Affiliation(s)
- Bi-Oh Park
- Incurable Disease Therapeutics Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; College of Pharmacy, Chungbuk National University, Cheongju, Republic of Korea
| | - Seong Heon Kim
- Incurable Disease Therapeutics Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; Department of Biomolecular Science, University of Science and Technology, Daejeon, Republic of Korea
| | - Gye Yeong Kong
- Incurable Disease Therapeutics Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; College of Pharmacy, Chungbuk National University, Cheongju, Republic of Korea
| | - Da Hui Kim
- Incurable Disease Therapeutics Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; College of Pharmacy, Chungbuk National University, Cheongju, Republic of Korea
| | - Mi So Kwon
- Next-generation Pharmaceutical Research Center, Korea Institute of Toxicology, Daejeon, Republic of Korea
| | - Su Ui Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | - Mun-Ock Kim
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | - Sungchan Cho
- Incurable Disease Therapeutics Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; Department of Biomolecular Science, University of Science and Technology, Daejeon, Republic of Korea
| | - Sangku Lee
- Incurable Disease Therapeutics Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | - Hyun-Jun Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy, Chungbuk National University, Cheongju, Republic of Korea
| | - Young Shin Kwak
- College of Pharmacy, Korea University, Sejong, Republic of Korea
| | - Sung Bae Lee
- Department of Brain Science, DGIST, Daegu, Republic of Korea.
| | - Sunhong Kim
- Incurable Disease Therapeutics Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Republic of Korea; Department of Biomolecular Science, University of Science and Technology, Daejeon, Republic of Korea.
| |
Collapse
|