751
|
Abstract
PURPOSE OF REVIEW The objective of this review is to critically assess the contributing role of the gut microbiota in human obesity and type 2 diabetes (T2D). RECENT FINDINGS Experiments in animal and human studies have produced growing evidence for the causality of the gut microbiome in developing obesity and T2D. The introduction of high-throughput sequencing technologies has provided novel insight into the interpersonal differences in microbiome composition and function. The intestinal microbiota is known to be associated with metabolic syndrome and related comorbidities. Associated diseases including obesity, T2D, and fatty liver disease (NAFLD/NASH) all seem to be linked to altered microbial composition; however, causality has not been proven yet. Elucidating the potential causal and personalized role of the human gut microbiota in obesity and T2D is highly prioritized.
Collapse
Affiliation(s)
- Ömrüm Aydin
- Department of Internal Medicine, MC Slotervaart, Amsterdam, The Netherlands
- Department of Internal Medicine, AMC-UVA, Amsterdam, The Netherlands
| | - Max Nieuwdorp
- Department of Internal Medicine, AMC-UVA, Amsterdam, The Netherlands
- Diabetes Center, Department of Internal Medicine, VU University Medical Center, Amsterdam, The Netherlands
- Wallenberg Laboratory, University of Gothenberg, Gothenberg, Sweden
| | - Victor Gerdes
- Department of Internal Medicine, MC Slotervaart, Amsterdam, The Netherlands.
- Department of Internal Medicine, AMC-UVA, Amsterdam, The Netherlands.
| |
Collapse
|
752
|
Abstract
PURPOSE OF REVIEW Bile acids act as activating signals of endogenous renal receptors: the nuclear receptor farnesoid X receptor (FXR) and the membrane-bound G protein-coupled bile acid receptor 1 (GPBAR1, also known as TGR5). In recent years, bile acids have emerged as important for renal pathophysiology by activating FXR and TGR5 and transcription factors relevant for lipid, cholesterol and carbohydrate metabolism, as well as genes involved in inflammation and renal fibrosis. RECENT FINDINGS Activation of bile acid receptors has a promising therapeutic potential in prevention of diabetic nephropathy and obesity-induced renal damage, as well as in nephrosclerosis. During the past decade, progress has been made in understanding the biology and mechanisms of bile acid receptors in the kidney and in the development of specific bile acid receptor agonists. SUMMARY In this review, we discuss current knowledge on the roles of FXR and TGR5 in the physiology of the kidney and the latest advances made in development and characterization of bile acid analogues that activate bile acid receptors for treatment of renal disease.
Collapse
|
753
|
GLP-2 receptor signaling controls circulating bile acid levels but not glucose homeostasis in Gcgr -/- mice and is dispensable for the metabolic benefits ensuing after vertical sleeve gastrectomy. Mol Metab 2018; 16:45-54. [PMID: 29937214 PMCID: PMC6157461 DOI: 10.1016/j.molmet.2018.06.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 06/04/2018] [Accepted: 06/06/2018] [Indexed: 02/06/2023] Open
Abstract
Objective Therapeutic interventions that improve glucose homeostasis such as attenuation of glucagon receptor (Gcgr) signaling and bariatric surgery share common metabolic features conserved in mice and humans. These include increased circulating levels of bile acids (BA) and the proglucagon-derived peptides (PGDPs), GLP-1 and GLP-2. Whether BA acting through TGR5 (Gpbar1) increases PGDP levels in these scenarios has not been examined. Furthermore, although the importance of GLP-1 action has been interrogated in Gcgr−/− mice and after bariatric surgery, whether GLP-2 contributes to the metabolic benefits of these interventions is not known. Methods To assess whether BA acting through Gpbar1 mediates improved glucose homeostasis in Gcgr−/− mice we generated and characterized Gcgr−/−:Gpbar1−/− mice. The contribution of GLP-2 receptor (GLP-2R) signaling to intestinal and metabolic adaptation arising following loss of the Gcgr was studied in Gcgr−/−:Glp2r−/− mice. The role of the GLP-2R in the metabolic improvements evident after bariatric surgery was studied in high fat-fed Glp2r−/− mice subjected to vertical sleeve gastrectomy (VSG). Results Circulating levels of BA were markedly elevated yet similar in Gcgr−/−:Gpbar1+/+ vs. Gcgr−/−:Gpbar1−/− mice. Loss of GLP-2R lowered levels of BA in Gcgr−/− mice. Gcgr−/−:Glp2r−/− mice also exhibited shifts in the proportion of circulating BA species. Loss of Gpbar1 did not impact body weight, intestinal mass, or glucose homeostasis in Gcgr−/− mice. In contrast, small bowel growth was attenuated in Gcgr−/−:Glp2r−/− mice. The improvement in glucose tolerance, elevated circulating levels of GLP-1, and glucose-stimulated insulin levels were not different in Gcgr−/−:Glp2r+/+ vs. Gcgr−/−:Glp2r−/− mice. Similarly, loss of the GLP-2R did not attenuate the extent of weight loss and improvement in glucose control after VSG. Conclusions These findings reveal that GLP-2R controls BA levels and relative proportions of BA species in Gcgr−/− mice. Nevertheless, the GLP-2R is not essential for i) control of body weight or glucose homeostasis in Gcgr−/− mice or ii) metabolic improvements arising after VSG in high fat-fed mice. Furthermore, despite elevations of circulating levels of BA, Gpbar1 does not mediate elevated levels of PGDPs or major metabolic phenotypes in Gcgr−/− mice. Collectively these findings refine our understanding of the relationship between Gpbar1, elevated levels of BA, PGDPs, and the GLP-2R in amelioration of metabolic derangements arising following loss of Gcgr signaling or after vertical sleeve gastrectomy. GLP-2 receptor controls bile acid levels in Gcgr−/− mice. Gpbar1 is not required for the metabolic benefits or elevated levels of PGDPs in Gcgr−/− mice. GLP-2 regulates gut adaptation in Gcgr−/− mice. Bile acid profiles are altered in Gcgr−/− mice following loss of GLP-2R. GLP-2R is not required for improvements in glucose homeostasis or weight loss after VSG in mice.
Collapse
|
754
|
Sheng L, Jena PK, Liu HX, Hu Y, Nagar N, Bronner DN, Settles ML, Bäumler AJ, Wan YJY. Obesity treatment by epigallocatechin-3-gallate-regulated bile acid signaling and its enriched Akkermansia muciniphila. FASEB J 2018; 32:fj201800370R. [PMID: 29882708 PMCID: PMC6219838 DOI: 10.1096/fj.201800370r] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 05/14/2018] [Indexed: 12/21/2022]
Abstract
Dysregulated bile acid (BA) synthesis is accompanied by dysbiosis, leading to compromised metabolism. This study analyzes the effect of epigallocatechin-3-gallate (EGCG) on diet-induced obesity through regulation of BA signaling and gut microbiota. The data revealed that EGCG effectively reduced diet-increased obesity, visceral fat, and insulin resistance. Gene profiling data showed that EGCG had a significant impact on regulating genes implicated in fatty acid uptake, adipogenesis, and metabolism in the adipose tissue. In addition, metabolomics analysis revealed that EGCG altered the lipid and sugar metabolic pathways. In the intestine, EGCG reduced the FXR agonist chenodeoxycholic acid, as well as the FXR-regulated pathway, suggesting intestinal FXR deactivation. However, in the liver, EGCG increased the concentration of FXR and TGR-5 agonists and their regulated signaling. Furthermore, our data suggested that EGCG activated Takeda G protein receptor (TGR)-5 based on increased GLP-1 release and elevated serum PYY level. EGCG and antibiotics had distinct antibacterial effects. They also differentially altered body weight and BA composition. EGCG, but not antibiotics, increased Verrucomicrobiaceae, under which EGCG promoted intestinal bloom of Akkermansia muciniphila. Excitingly, A. muciniphila was as effective as EGCG in treating diet-induced obesity. Together, EGCG shifts gut microbiota and regulates BA signaling thereby having a metabolic beneficial effect.-Sheng, L., Jena, P. K., Liu, H.-X., Hu, Y., Nagar, N., Bronner, D. N., Settles, M. L., Bäumler, A. J. Wan, Y.-J. Y. Obesity treatment by epigallocatechin-3-gallate-regulated bile acid signaling and its enriched Akkermansia muciniphila.
Collapse
Affiliation(s)
- Lili Sheng
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, California, USA
| | - Prasant Kumar Jena
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, California, USA
| | - Hui-Xin Liu
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, California, USA
| | - Ying Hu
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, California, USA
| | - Nidhi Nagar
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, California, USA
| | - Denise N. Bronner
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, California, USA
| | - Matthew L. Settles
- Bioinformatics Core Facility in the Genome Center, University of California, Davis, Davis, California, USA
| | - Andreas J. Bäumler
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, California, USA
| | - Yu-Jui Yvonne Wan
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, California, USA
| |
Collapse
|
755
|
Zhai H, Li Z, Peng M, Huang Z, Qin T, Chen L, Li H, Zhang H, Zhang W, Xu G. Takeda G Protein-Coupled Receptor 5-Mechanistic Target of Rapamycin Complex 1 Signaling Contributes to the Increment of Glucagon-Like Peptide-1 Production after Roux-en-Y Gastric Bypass. EBioMedicine 2018; 32:201-214. [PMID: 29859856 PMCID: PMC6020750 DOI: 10.1016/j.ebiom.2018.05.026] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 05/20/2018] [Accepted: 05/21/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The mechanism by which Roux-en-Y Gastric Bypass (RYGB) increases the secretion of glucagon-like peptide-1 (GLP-1) remains incompletely defined. Here we investigated whether TGR5-mTORC1 signaling mediates the RYGB-induced alteration in GLP-1 production in mice and human beings. METHODS Circulating bile acids, TGR5-mTORC1 signaling, GLP-1 synthesis and secretion were determined in lean or obese male C57BL/6 mice with or without RYGB operation, as well as in normal glycemic subjects, obese patients with type 2 diabetes before and after RYGB. RESULTS Positive relationships were observed among circulating bile acids, ileal mechanistic target of rapamycin complex 1 (mTORC1) signaling and GLP-1 during changes in energy status in the present study. RYGB increased circulating bile acids, ileal Takeda G protein-coupled receptor 5 (TGR5) and mTORC1 signaling activity, as well as GLP-1 production in both mice and human subjects. Inhibition of ileal mTORC1 signaling by rapamycin significantly attenuated the stimulation of bile acid secretion, TGR5 expression and GLP-1 synthesis induced by RYGB in lean and diet-induced obese mice. GLP-1 production and ileal TGR5-mTORC1 signaling were positively correlated with plasma deoxycholic acid (DCA) in mice. Treatment of STC-1 cells with DCA stimulated the production of GLP-1. This effect was associated with a significant enhancement of TGR5-mTORC1 signaling. siRNA knockdown of mTORC1 or TGR5 abolished the enhancement of GLP-1 synthesis induced by DCA. DCA increased interaction between mTOR-regulatory-associated protein of mechanistic target of rapamycin (Raptor) and TGR5 in STC-1 cells. INTERPRETATION Deoxycholic acid-TGR5-mTORC1 signaling contributes to the up-regulation of GLP-1 production after RYGB.
Collapse
Affiliation(s)
- Hening Zhai
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China; Endoscopy Center, The First Affiliated Hospital of Jinan University, 613 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510630, China
| | - Zhi Li
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Miao Peng
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Zhaoqi Huang
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Tingfeng Qin
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Linxi Chen
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Hanbing Li
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Heng Zhang
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Weizhen Zhang
- Shenzhen University Diabetes Center, Shenzhen University Health Science Center, Shenzhen, Guangdong 518060, China; Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI 48109-0346, USA.
| | - Geyang Xu
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China.
| |
Collapse
|
756
|
Abstract
The gut microbiota has been recognized as an important factor in the development of metabolic diseases such as obesity and is considered an endocrine organ involved in the maintenance of energy homeostasis and host immunity. Dysbiosis can change the functioning of the intestinal barrier and the gut-associated lymphoid tissues (GALT) by allowing the passage of structural components of bacteria, such as lipopolysaccharides (LPS), which activate inflammatory pathways that may contribute to the development of insulin resistance. Furthermore, intestinal dysbiosis can alter the production of gastrointestinal peptides related to satiety, resulting in an increased food intake. In obese people, this dysbiosis seems be related to increases of the phylum Firmicutes, the genus Clostridium, and the species Eubacterium rectale, Clostridium coccoides, Lactobacillus reuteri, Akkermansia muciniphila, Clostridium histolyticum, and Staphylococcus aureus.
Collapse
Affiliation(s)
- Aline Corado Gomes
- Clinical and Sports Nutrition Research Laboratory (LABINCE), Faculty of Nutrition, Goiás Federal University, Goiânia, Goiás, Brazil,CONTACT Dra. Aline Corado Gomes Clinical and Sports Nutrition Research Laboratory (LABINCE), Faculty of Nutrition, Goiás Federal University, Setor Leste Universitário, Goiânia, St. 227, Block 68, Goiânia GO, Brazil
| | - Christian Hoffmann
- Department of Food Sciences and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - João Felipe Mota
- Clinical and Sports Nutrition Research Laboratory (LABINCE), Faculty of Nutrition, Goiás Federal University, Goiânia, Goiás, Brazil
| |
Collapse
|
757
|
Abstract
Bile acids facilitate intestinal nutrient absorption and biliary cholesterol secretion to maintain bile acid homeostasis, which is essential for protecting liver and other tissues and cells from cholesterol and bile acid toxicity. Bile acid metabolism is tightly regulated by bile acid synthesis in the liver and bile acid biotransformation in the intestine. Bile acids are endogenous ligands that activate a complex network of nuclear receptor farnesoid X receptor and membrane G protein-coupled bile acid receptor-1 to regulate hepatic lipid and glucose metabolic homeostasis and energy metabolism. The gut-to-liver axis plays a critical role in the regulation of enterohepatic circulation of bile acids, bile acid pool size, and bile acid composition. Bile acids control gut bacteria overgrowth, and gut bacteria metabolize bile acids to regulate host metabolism. Alteration of bile acid metabolism by high-fat diets, sleep disruption, alcohol, and drugs reshapes gut microbiome and causes dysbiosis, obesity, and metabolic disorders. Gender differences in bile acid metabolism, FXR signaling, and gut microbiota have been linked to higher prevalence of fatty liver disease and hepatocellular carcinoma in males. Alteration of bile acid homeostasis contributes to cholestatic liver diseases, inflammatory diseases in the digestive system, obesity, and diabetes. Bile acid-activated receptors are potential therapeutic targets for developing drugs to treat metabolic disorders.
Collapse
Affiliation(s)
- John Y. L. Chiang
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Jessica M. Ferrell
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| |
Collapse
|
758
|
Sasaki T, Kuboyama A, Mita M, Murata S, Shimizu M, Inoue J, Mori K, Sato R. The exercise-inducible bile acid receptor Tgr5 improves skeletal muscle function in mice. J Biol Chem 2018; 293:10322-10332. [PMID: 29773650 DOI: 10.1074/jbc.ra118.002733] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 05/15/2018] [Indexed: 12/15/2022] Open
Abstract
TGR5 (also known as G protein-coupled bile acid receptor 1, GPBAR1) is a G protein-coupled bile acid receptor that is expressed in many diverse tissues. TGR5 is involved in various metabolic processes, including glucose metabolism and energy expenditure; however, TGR5's function in skeletal muscle is not fully understood. Using both gain- and loss-of-function mouse models, we demonstrate here that Tgr5 activation promotes muscle cell differentiation and muscle hypertrophy. Both young and old transgenic mice with muscle-specific Tgr5 expression exhibited increased muscle strength. Moreover, we found that Tgr5 expression is increased by the unfolded protein response (UPR), which is an adaptive response required for maintenance of endoplasmic reticulum (ER) homeostasis. Both ER stress response element (ERSE)- and unfolded protein response element (UPRE)-like sites are present in the 5' upstream region of the Tgr5 gene promoter and are essential for Tgr5 expression by Atf6α (activating transcription factor 6α), a well known UPR-activated transcriptional regulator. We observed that in the skeletal muscle of mice, exercise-induced UPR increases Tgr5 expression, an effect that was abrogated in Atf6α KO mice, indicating that Atf6α is essential for this response. These findings indicate that the bile acid receptor Tgr5 contributes to improved muscle function and provide an additional explanation for the beneficial effects of exercise on skeletal muscle activity.
Collapse
Affiliation(s)
| | | | - Moeko Mita
- From the Food Biochemistry Laboratory and
| | | | | | - Jun Inoue
- From the Food Biochemistry Laboratory and
| | - Kazutoshi Mori
- the Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan, and
| | - Ryuichiro Sato
- From the Food Biochemistry Laboratory and .,Nutri-Life Science Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo 113-8657, Japan.,AMED-CREST, Japan Agency for Medical Research and Development, Tokyo 100-0004, Japan
| |
Collapse
|
759
|
Hylemon PB, Harris SC, Ridlon JM. Metabolism of hydrogen gases and bile acids in the gut microbiome. FEBS Lett 2018; 592:2070-2082. [PMID: 29683480 DOI: 10.1002/1873-3468.13064] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/04/2018] [Accepted: 04/10/2018] [Indexed: 12/26/2022]
Abstract
The human gut microbiome refers to a highly diverse microbial ecosystem, which has a symbiotic relationship with the host. Molecular hydrogen (H2 ) and carbon dioxide (CO2 ) are generated by fermentative metabolism in anaerobic ecosystems. H2 generation and oxidation coupled to CO2 reduction to methane or acetate help maintain the structure of the gut microbiome. Bile acids are synthesized by hepatocytes from cholesterol in the liver and are important regulators of host metabolism. In this Review, we discuss how gut bacteria metabolize hydrogen gases and bile acids in the intestinal tract and the consequences on host physiology. Finally, we focus on bile acid metabolism by the Actinobacterium Eggerthella lenta. Eggerthella lenta appears to couple hydroxyl group oxidations to reductive acetogenesis under a CO2 or N2 atmosphere, but not under H2 . Hence, at low H2 levels, E. lenta is proposed to use NADH from bile acid hydroxyl group oxidations to reduce CO2 to acetate.
Collapse
Affiliation(s)
- Phillip B Hylemon
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA, USA.,McGuire Veterans Hospital, Richmond, VA, USA
| | - Spencer C Harris
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA, USA.,McGuire Veterans Hospital, Richmond, VA, USA
| | - Jason M Ridlon
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, IL, USA
| |
Collapse
|
760
|
Zhao LJ, Zhang SF. Activation of TGR5 promotes mitochondrial biogenesis in human aortic endothelial cells. Biochem Biophys Res Commun 2018; 500:952-957. [PMID: 29709472 DOI: 10.1016/j.bbrc.2018.04.210] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 04/26/2018] [Indexed: 01/22/2023]
Abstract
Impairment of mitochondrial biogenesis has been associated with vascular pathophysiology. The G-protein-coupled receptor (TGR5) is an important mediator of bile acid signaling and glucose metabolism. However, the effects of TGR5 on mitochondrial biogenesis in endothelial cells remain elusive. In this study, we found that activation of TGR5 using its specific agonist taurolithocholic acid (TLCA) promoted the expression of PGC-1α, a master regulator of mitochondrial biogenesis in human aortic endothelial cells (HAECs). Additionally, activation of TGR5 increased the expression of PGC-1α target genes, such as NRF1 and TFAM. Indeed, we found that TLCA treatment promoted mitochondrial biogenesis by increasing mitochondrial mass, mitochondrial-to-nuclear DNA (mtDNA/nDNA), COX-Ⅰ expression, and cytochrome c oxidase activity in HAECs. Notably, our results displayed that activation of TGR5 resulted in a functional gain in mitochondria by increasing the rate of respiration and ATP production. Mechanistically, we found that TLCA treatment activated the transcriptional factor CREB by inducing the phosphorylation of CREB at Ser133. Using the PKA/CREB inhibitor H89 abolished the effects of TLCA on PGC-1α, NRF1 and TFAM expression as well as the increase in mtDNA/nDNA and ATP production. These findings suggest that activation of TGR5 promoted mitochondrial biogenesis in endothelial cells, which is mediated by the CREB/PGC-1α signaling pathway.
Collapse
Affiliation(s)
- Li-Jun Zhao
- Department of Internal Medicine, West China School of Medicine, Sichuan University, Chengdu 610041, Sichuan Province, China.
| | - Shi-Fang Zhang
- Department of Pulmonary Disease, Institute of Respiratory Disease, Chengdu Second People's Hospital, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
761
|
Hu X, Chandler JD, Orr ML, Hao L, Liu K, Uppal K, Go YM, Jones DP. Selenium Supplementation Alters Hepatic Energy and Fatty Acid Metabolism in Mice. J Nutr 2018; 148:675-684. [PMID: 29982657 PMCID: PMC6454983 DOI: 10.1093/jn/nxy036] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 11/16/2017] [Accepted: 02/09/2018] [Indexed: 12/11/2022] Open
Abstract
Background Human and animal studies have raised concerns that supplemental selenium can increase the risk of metabolic disorders, but underlying mechanisms are unclear. Objective We used an integrated transcriptome and metabolome analysis of liver to test for functional pathway and network responses to supplemental selenium in mice. Methods Male mice (8-wk-old, C57BL/6J) fed a standard diet (0.41 ppm Se) were given selenium (Na2SeO4, 20 μmol/L) or vehicle (drinking water) for 16 wk. Livers were analyzed for selenium concentration, activity of selenoproteins, reduced glutathione (GSH) redox state, gene expression, and high-resolution metabolomics. Transcriptomic and nontargeted metabolomic data were analyzed with biostatistics, bioinformatics, pathway enrichment analysis, and combined transcriptome-metabolome-wide association study (TMWAS). Results Mice supplemented with selenium had greater body mass gain from baseline to 16 wk (55% ± 5%) compared with controls (40% ± 3%) (P < 0.05); however, no difference was observed in liver selenium content, selenoenzyme transcripts, or enzyme activity. Selenium was higher in the heart, kidney, and urine of mice supplemented with selenium. Gene enrichment analysis showed that supplemental selenium altered pathways of lipid and energy metabolism. Integrated transcriptome and metabolome network analysis showed 2 major gene-metabolite clusters, 1 centered on the transcript for the bidirectional glucose transporter 2 (Glut2) and the other centered on the transcripts for carnitine-palmitoyl transferase 2 (Cpt2) and acetyl-CoA acyltransferase (Acaa1). Pathway analysis showed that highly associated metabolites (P < 0.05) were enriched in fatty acid metabolism and bile acid biosynthesis, including acylcarnitines, triglycerides and glycerophospholipids, long-chain acyl-coenzyme As, phosphatidylcholines, and sterols. TMWAS of body weight gain confirmed changes in the same pathways. Conclusions Supplemental selenium in mice alters hepatic fatty acid and energy metabolism and causes increases in body mass. A lack of effect on hepatic selenium content suggests that signaling involves an extrahepatic mechanism.
Collapse
Affiliation(s)
- Xin Hu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA
| | - Joshua D Chandler
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA
| | - Michael L Orr
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA
| | - Li Hao
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA
| | - Ken Liu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA
| | - Karan Uppal
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA
| | - Young-Mi Go
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA
| | - Dean P Jones
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA
| |
Collapse
|
762
|
Wei M, Shao Y, Liu QR, Wu QZ, Zhang X, Zhong MW, Liu SZ, Zhang GY, Hu SY. Bile acid profiles within the enterohepatic circulation in a diabetic rat model after bariatric surgeries. Am J Physiol Gastrointest Liver Physiol 2018; 314:G537-G546. [PMID: 29351394 DOI: 10.1152/ajpgi.00311.2017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Bile acids (BAs), which are synthesized in the liver and cycled in the enterohepatic circulation, have been recognized as signaling molecules by activating their receptors in the intestine and liver. Serum taurine-conjugated BAs have been shown to be elevated after bariatric surgeries although the postoperative BA profiles within the enterohepatic circulation have not been investigated. Clarification of these profiles could help explain the mechanisms by which bariatric surgery leads to BA profile alterations and subsequent metabolic effects. We performed duodenal-jejunal bypass (DJB), sleeve gastrectomy (SG), and sham procedures in an obese diabetic rat model induced by high-fat diet and streptozotocin. The weight loss and antidiabetic effects were evaluated postsurgery. BA profiles in the systemic serum and within the enterohepatic circulation were analyzed, together with the expression of related BA transporters and enzymes at week 12 after surgery. Compared with sham, SG induced sustained weight loss, and both DJB and SG significantly improved glucose tolerance and insulin sensitivity with enhanced glucagon-like peptide 1 secretion. Similar to changes in the serum, BAs, especially taurine-conjugated species, were also elevated in the enterohepatic circulation (bile and portal vein) after DJB and SG. In addition, the expression of key BA transporters and conjugational enzymes was elevated postoperatively, whereas the enzymes responsible for BA synthesis were decreased. In conclusion, DJB and SG elevated BA levels in the systemic serum and enterohepatic circulation, especially taurine-conjugated species, which likely indicates increased ileal reabsorption and hepatic conjugation rather than synthesis. NEW & NOTEWORTHY Bile acids (BAs) have been implicated as potential mediators of the weight-independent effects of bariatric surgery. For the first time, we discovered that duodenal-jejunal bypass and sleeve gastrectomy elevated BAs, particularly the taurine-conjugated species in the enterohepatic circulation, likely through the promotion of ileal reabsorption and hepatic conjugation rather than BA synthesis. These findings will improve our understanding of BA metabolism after bariatric surgery and their subsequent metabolic effects.
Collapse
Affiliation(s)
- Meng Wei
- Department of General Surgery, Qilu Hospital of Shandong University , Jinan , People's Republic of China
| | - Yi Shao
- Department of General Surgery, Qilu Hospital of Shandong University , Jinan , People's Republic of China
| | - Qiao-Ran Liu
- Department of General Surgery, Qilu Hospital of Shandong University , Jinan , People's Republic of China
| | - Qun-Zheng Wu
- Department of General Surgery, Qilu Hospital of Shandong University , Jinan , People's Republic of China
| | - Xiang Zhang
- Department of General Surgery, Qilu Hospital of Shandong University , Jinan , People's Republic of China
| | - Ming-Wei Zhong
- Department of General Surgery, Qilu Hospital of Shandong University , Jinan , People's Republic of China
| | - Shao-Zhuang Liu
- Department of General Surgery, Qilu Hospital of Shandong University , Jinan , People's Republic of China
| | - Guang-Yong Zhang
- Department of General Surgery, Qilu Hospital of Shandong University , Jinan , People's Republic of China
| | - San-Yuan Hu
- Department of General Surgery, Qilu Hospital of Shandong University , Jinan , People's Republic of China
| |
Collapse
|
763
|
Minnifield BA, Aslibekyan SW. The Interplay Between the Microbiome and Cardiovascular Risk. CURRENT GENETIC MEDICINE REPORTS 2018. [DOI: 10.1007/s40142-018-0142-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
764
|
Jena PK, Sheng L, Nagar N, Wu C, Barile D, Mills DA, Wan YJY. Synbiotics Bifidobacterium infantis and milk oligosaccharides are effective in reversing cancer-prone nonalcoholic steatohepatitis using western diet-fed FXR knockout mouse models. J Nutr Biochem 2018; 57:246-254. [PMID: 29800811 DOI: 10.1016/j.jnutbio.2018.04.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 04/12/2018] [Accepted: 04/17/2018] [Indexed: 12/23/2022]
Abstract
Milk oligosaccharides (MO) selectively increase the growth of Bifidobacterium infantis (B. infantis). This study examines the effects of bovine MO and B. infantis in preventing nonalcoholic steatohepatitis (NASH) in Western diet (WD)-fed bile acid (BA) receptor FXR (farnesoid x receptor) knockout (KO) mice. WD-fed FXR KO mice have cancer-prone NASH and reduced B. infantis. MO and/or B. infantis supplementation improved their insulin sensitivity and reduced hepatic inflammation. Additionally, B. infantis, but not MO, decreased hepatic triglyceride and cholesterol. A combination of both further reduced hepatic cholesterol, the precursor of BAs. All three treatments modulated serum and hepatic BA profile. Moreover, B. infantis and MO decreased hepatic CYP7A1 and induced Sult2a1, Sult2a2, and Sult2a3 suggesting reduced BA synthesis and increased detoxification. Furthermore, B. infantis and MO increased ileal BA membrane receptor TGR5-regulated signaling. Together, via BA-regulated signaling, synbiotics B. infantis and MO have their unique and combined effects in reversing NASH.
Collapse
Affiliation(s)
- Prasant Kumar Jena
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento 95817, CA, USA
| | - Lili Sheng
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento 95817, CA, USA
| | - Nidhi Nagar
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento 95817, CA, USA
| | - Chao Wu
- Research and Development, Hilmar Ingredients, Hilmar 95324, CA, USA
| | - Daniela Barile
- Department of Food Science and Technology, University of California, Davis 95616, CA, USA
| | - David A Mills
- Department of Food Science and Technology, University of California, Davis 95616, CA, USA; Department of Viticulture and Enology, University of California, Davis 95616, CA, USA
| | - Yui-Jui Yvonne Wan
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento 95817, CA, USA.
| |
Collapse
|
765
|
Yu Q, Jiang Z, Zhang L. Bile acid regulation: A novel therapeutic strategy in non-alcoholic fatty liver disease. Pharmacol Ther 2018; 190:81-90. [PMID: 29684468 DOI: 10.1016/j.pharmthera.2018.04.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is characterized by excessive fat deposition in the liver in the absence of significant alcohol consumption. Dysregulated bile acid (BA) metabolism is an important indicator in the pathology of NAFLD, which could progress into more severe forms of liver injury. Lipid metabolism, immune environment and intestinal bacteria are all affected by dysregulated BA metabolism directly, but the mechanisms remain unclear. Several drug candidates that target BA metabolism, either used alone or in combination with other agents, are currently under development for treatment of NAFLD. Here, we summarize the relationship of dysregulated BA metabolism and NAFLD, discuss the effects and mechanisms of dysregulated BAs-induced lipid metabolism disorder. Challenges in developing novel treatments are also discussed.
Collapse
Affiliation(s)
- Qinwei Yu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Zhenzhou Jiang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China.
| | - Luyong Zhang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Center for Drug Screening and Pharmacodynamics Evaluation, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
766
|
Wang Y, Li J, Matye D, Zhang Y, Dennis K, Ding WX, Li T. Bile acids regulate cysteine catabolism and glutathione regeneration to modulate hepatic sensitivity to oxidative injury. JCI Insight 2018; 3:99676. [PMID: 29669937 DOI: 10.1172/jci.insight.99676] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 03/20/2018] [Indexed: 12/23/2022] Open
Abstract
Bile acids are signaling molecules that critically control hepatocellular function. Disrupted bile acid homeostasis may be implicated in the pathogenesis of chronic liver diseases. Glutathione is an important antioxidant that protects the liver against oxidative injury. Various forms of liver disease share the common characteristics of reduced cellular glutathione and elevated oxidative stress. This study reports a potentially novel physiological function of bile acids in regulating hepatic sulfur amino acid and glutathione metabolism. We found that bile acids strongly inhibited the cysteine dioxygenase type-1-mediated (CDO1-mediated) cysteine catabolic pathway via a farnesoid X receptor-dependent mechanism. Attenuating this bile acid repressive effect depleted the free cysteine pool and reduced the glutathione concentration in mouse liver. Upon acetaminophen challenge, cholestyramine-fed mice showed impaired hepatic glutathione regeneration capacity and markedly worsened liver injury, which was fully prevented by N-acetylcysteine administration. These effects were recapitulated in CDO1-overexpressing hepatocytes. Findings from this study support the importance of maintaining bile acid homeostasis under physiological and pathophysiological conditions, as altered hepatic bile acid signaling may negatively impact the antioxidant defense mechanism and sensitivity to oxidative injury. Furthermore, this finding provides a possible explanation for the reported mild hepatotoxicity associated with the clinical use of bile acid sequestrants in human patients.
Collapse
Affiliation(s)
- Yifeng Wang
- Department of Pharmacology, Toxicology and Therapeutics, and
| | - Jibiao Li
- Department of Pharmacology, Toxicology and Therapeutics, and
| | - David Matye
- Department of Pharmacology, Toxicology and Therapeutics, and
| | - Yuxia Zhang
- Department of Pharmacology, Toxicology and Therapeutics, and
| | - Katie Dennis
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, and
| | - Tiangang Li
- Department of Pharmacology, Toxicology and Therapeutics, and
| |
Collapse
|
767
|
Sepe V, Distrutti E, Fiorucci S, Zampella A. Farnesoid X receptor modulators 2014-present: a patent review. Expert Opin Ther Pat 2018; 28:351-364. [DOI: 10.1080/13543776.2018.1459569] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Valentina Sepe
- Department of Pharmacy, University of Naples “Federico II”, Naples, Italy
| | | | - Stefano Fiorucci
- Department of Experimental and Clinical Medicine, University of Perugia, Perugia, Italy
| | - Angela Zampella
- Department of Pharmacy, University of Naples “Federico II”, Naples, Italy
| |
Collapse
|
768
|
Martin CR, Osadchiy V, Kalani A, Mayer EA. The Brain-Gut-Microbiome Axis. Cell Mol Gastroenterol Hepatol 2018; 6:133-148. [PMID: 30023410 PMCID: PMC6047317 DOI: 10.1016/j.jcmgh.2018.04.003] [Citation(s) in RCA: 776] [Impact Index Per Article: 110.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 04/04/2018] [Indexed: 12/12/2022]
Abstract
Preclinical and clinical studies have shown bidirectional interactions within the brain-gut-microbiome axis. Gut microbes communicate to the central nervous system through at least 3 parallel and interacting channels involving nervous, endocrine, and immune signaling mechanisms. The brain can affect the community structure and function of the gut microbiota through the autonomic nervous system, by modulating regional gut motility, intestinal transit and secretion, and gut permeability, and potentially through the luminal secretion of hormones that directly modulate microbial gene expression. A systems biological model is proposed that posits circular communication loops amid the brain, gut, and gut microbiome, and in which perturbation at any level can propagate dysregulation throughout the circuit. A series of largely preclinical observations implicates alterations in brain-gut-microbiome communication in the pathogenesis and pathophysiology of irritable bowel syndrome, obesity, and several psychiatric and neurologic disorders. Continued research holds the promise of identifying novel therapeutic targets and developing treatment strategies to address some of the most debilitating, costly, and poorly understood diseases.
Collapse
Key Words
- 2BA, secondary bile acid
- 5-HT, serotonin
- ANS, autonomic nervous system
- ASD, autism spectrum disorder
- BBB, blood-brain barrier
- BGM, brain-gut-microbiome
- CNS, central nervous system
- ECC, enterochromaffin cell
- EEC, enteroendocrine cell
- FFAR, free fatty acid receptor
- FGF, fibroblast growth factor
- FXR, farnesoid X receptor
- GF, germ-free
- GI, gastrointestinal
- GLP-1, glucagon-like peptide-1
- GPR, G-protein–coupled receptor
- IBS, irritable bowel syndrome
- Intestinal Permeability
- Irritable Bowel Syndrome
- LPS, lipopolysaccharide
- SCFA, short-chain fatty acid
- SPF, specific-pathogen-free
- Serotonin
- Stress
- TGR5, G protein-coupled bile acid receptor
- Trp, tryptophan
Collapse
Affiliation(s)
| | | | | | - Emeran A. Mayer
- Correspondence Address correspondence to: Emeran A. Mayer, MD, G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California at Los Angeles, MC737818-10833 Le Conte Avenue, Los Angeles, California 90095-7378. fax: (310) 825-1919.
| |
Collapse
|
769
|
Ong HS, Yim HCH. Microbial Factors in Inflammatory Diseases and Cancers. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1024:153-174. [PMID: 28921469 DOI: 10.1007/978-981-10-5987-2_7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The intestinal microbes form a symbiotic relationship with their human host to harvest energy for themselves and their host and to shape the immune system of their host. However, alteration of this relationship, which is named as a dysbiosis, has been associated with the development of different inflammatory diseases and cancers. It is found that metabolites, cellular components, and virulence factors derived from the gut microbiota interact with the host locally or systemically to modulate the dysbiosis and the development of these diseases. In this book chapter, we discuss the role of these microbial factors in regulating the host signaling pathways, the composition and load of the gut microbiota, the co-metabolism of the host and the microbiota, the host immune system, and physiology. In particular, we highlight how each microbial factor can contribute in the manifestation of many diseases such as cancers, Inflammatory Bowel Diseases, obesity, type-2 diabetes, non-alcoholic fatty liver diseases, nonalcoholic steatohepatitis, and cardiovascular diseases.
Collapse
Affiliation(s)
- Hong Sheng Ong
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Building 131, Garran Road, Acton, Canberra, 2601, ACT, Australia
| | - Howard Chi Ho Yim
- Department of Medicine, St George & Sutherland Clinical School, Faculty of Medicine, University of New South Wales, Australia, Level 2 Clinical Sciences (WR Pitney) Building, St George Hospital, Short St, Kogarah, NSW, 2217, Australia.
| |
Collapse
|
770
|
Evaluation of novel TGR5 agonist in combination with Sitagliptin for possible treatment of type 2 diabetes. Bioorg Med Chem Lett 2018; 28:1849-1852. [PMID: 29655980 DOI: 10.1016/j.bmcl.2018.04.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 03/31/2018] [Accepted: 04/04/2018] [Indexed: 12/12/2022]
Abstract
TGR5 is a member of G protein-coupled receptor (GPCR) superfamily, a promising molecular target for metabolic diseases. Activation of TGR5 promotes secretion of glucagon-like peptide-1 (GLP-1), which activates insulin secretion. A series of 2-thio-imidazole derivatives have been identified as novel, potent and orally efficacious TGR5 agonists. Compound 4d, a novel TGR5 agonist, in combination with Sitagliptin, a DPP-4 inhibitor, has demonstrated an adequate GLP-1 secretion and glucose lowering effect in animal models, suggesting a potential clinical option in treatment of type-2 diabetes.
Collapse
|
771
|
Zhao CL, Amin A, Hui Y, Yang D, Cao W. TGR5 expression in normal kidney and renal neoplasms. Diagn Pathol 2018; 13:22. [PMID: 29606134 PMCID: PMC5880016 DOI: 10.1186/s13000-018-0700-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 03/20/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The G protein-coupled bile acid receptor (TGR5) is a cell surface receptor which induces the production of intracellular cAMP and promotes epithelial-mesenchymal transition in gastric cancer cell lines. TGR5 is found in a wide variety of tissues including the kidney. However, the patterns of TGR5 expression have not been well characterized in physiologic kidney or renal neoplasms. We explore the expression of TGR5 in benign renal tissue and renal neoplasms and assess its utility as a diagnostic marker. METHODS Sixty-one renal cortical neoplasms from 2000 to 2014 were retrieved. TGR5 protein expression was examined by immunohistochemistry. TGR5 mRNA was also measured by real-time PCR. RESULTS In normal renal tissue, TGR5 was strongly positive in collecting ducts, distal convoluted tubules and thin loop of Henle. Proximal convoluted tubules showed absent or focal weak staining. In clear cell renal cell carcinomas (RCCs), 25 of 27 cases (92%) were negative for TGR5 (p < 0.001). TGR5 mRNA was also significantly decreased in clear cell RCCs, suggesting that decreased TGR5 protein expression may be attributable to the downregulation of TGR5 mRNA in these tumors. All 11 papillary RCCs expressed TGR5 with 45% (5/11) exhibiting moderate to strong staining. All chromophobe RCCs and oncocytomas were positive for TGR5 with weak to moderate staining. TGR5 mRNA expression in these tumors was similar to normal kidney. All urothelial carcinomas of the renal pelvis strongly expressed TGR5 including a poorly differentiated urothelial carcinoma with sarcomatoid features. CONCLUSION TGR5 is strongly expressed in collecting ducts, distal convoluted tubules and thin loop of Henle. TGR5 protein and mRNA expression were notably decreased in clear cell RCCs and may be helpful in differentiating these tumors from other RCCs.
Collapse
Affiliation(s)
- Chaohui Lisa Zhao
- Department of Pathology, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, 593 Eddy Street, APC 12, Providence, RI, 02903, USA
| | - Ali Amin
- Department of Pathology, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, 593 Eddy Street, APC 12, Providence, RI, 02903, USA
| | - Yiang Hui
- Department of Pathology, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, 593 Eddy Street, APC 12, Providence, RI, 02903, USA
| | - Dongfang Yang
- Department of Pathology, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, 593 Eddy Street, APC 12, Providence, RI, 02903, USA
| | - Weibiao Cao
- Department of Pathology, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, 593 Eddy Street, APC 12, Providence, RI, 02903, USA. .,Department of Medicine, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, 02903, USA.
| |
Collapse
|
772
|
de Faria Ghetti F, Oliveira DG, de Oliveira JM, de Castro Ferreira LEVV, Cesar DE, Moreira APB. Influence of gut microbiota on the development and progression of nonalcoholic steatohepatitis. Eur J Nutr 2018; 57:861-876. [PMID: 28875318 DOI: 10.1007/s00394-017-1524-x] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 08/06/2017] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Nonalcoholic steatohepatitis (NASH) is characterized by the presence of steatosis, inflammation, and ballooning degeneration of hepatocytes, with or without fibrosis. The prevalence of NASH has increased with the obesity epidemic, but its etiology is multifactorial. The current studies suggest the role of gut microbiota in the development and progression of NASH. The aim is to review the studies that investigate the relationship between gut microbiota and NASH. These review also discusses the pathophysiological mechanisms and the influence of diet on the gut-liver axis. RESULT The available literature has proposed mechanisms for an association between gut microbiota and NASH, such as: modification energy homeostasis, lipopolysaccharides (LPS)-endotoxemia, increased endogenous production of ethanol, and alteration in the metabolism of bile acid and choline. There is evidence to suggest that NASH patients have a higher prevalence of bacterial overgrowth in the small intestine and changes in the composition of the gut microbiota. However, there is still a controversy regarding the microbiome profile in this population. The abundance of Bacteroidetes phylum may be increased, decreased, or unaltered in NASH patients. There is an increase in the Escherichia and Bacteroides genus. There is depletion of certain taxa, such as Prevotella and Faecalibacterium. CONCLUSION Although few studies have evaluated the composition of the gut microbiota in patients with NASH, it is observed that these individuals have a distinct gut microbiota, compared to the control groups, which explains, at least in part, the genesis and progression of the disease through multiple mechanisms. Modulation of the gut microbiota through diet control offers new challenges for future studies.
Collapse
Affiliation(s)
- Fabiana de Faria Ghetti
- Universitary Hospital and School of Medicine, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil.
- Unidade de Nutrição Clínica, Hospital Universitário, Rua Catulo Breviglieri, s/n, Bairro Santa Catarina, Juiz de Fora, Minas Gerais, CEP 36036-330, Brazil.
| | - Daiane Gonçalves Oliveira
- Universitary Hospital and School of Medicine, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Juliano Machado de Oliveira
- Universitary Hospital and School of Medicine, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | | | | | | |
Collapse
|
773
|
Ippagunta SM, Kharitonenkov A, Adams AC, Hillgartner FB. Cholic Acid Supplementation of a High-Fat Obesogenic Diet Suppresses Hepatic Triacylglycerol Accumulation in Mice via a Fibroblast Growth Factor 21-Dependent Mechanism. J Nutr 2018; 148:510-517. [PMID: 29659970 DOI: 10.1093/jn/nxy022] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 01/25/2018] [Indexed: 12/13/2022] Open
Abstract
Background Supplementation of a high-fat obesogenic diet (HFD) with cholic acid (CA) suppresses the development of obesity, insulin resistance, and hepatic steatosis in mice. Objective We investigated the role of fibroblast growth factor 21 (FGF21) in mediating the beneficial actions of CA on metabolic syndrome. Methods Male 7-wk-old wild-type (WT) mice and FGF21 knockout (FGF21KO) mice were fed an HFD for 12 wk followed by a 4-wk period in which the mice were fed the HFD alone or supplemented with 0.5% CA. Body composition, gross energy efficiency, glucose tolerance, homeostasis model assessment of insulin resistance (HOMA-IR), and hepatic triacylglycerol (TG) concentrations were measured. Results CA administration improved glucose tolerance and decreased total body fat accretion, gross energy efficiency, fasting blood glucose concentrations, and HOMA-IR in both WT mice and FGF21KO mice. The extent of the effect of CA on glucose tolerance, fasting blood glucose concentrations, and HOMA-IR was similar in both mouse strains, whereas the extent of the effect of CA on total body fat accretion and gross energy efficiency was 4.2- to 4.4-fold greater in FGF21KO mice than in WT mice. Further analyses showed that CA decreased hepatic TG concentrations in WT mice (49%) but had no effect on hepatic TG concentrations in FGF21KO mice. CA decreased the activation state of hepatic acetyl-CoA carboxylase 1 (ACC1) and adipose tissue hormone-sensitive lipase (HSL) in WT mice but was not effective in decreasing the activation of ACC1 and HSL in FGF21KO mice. Conclusions FGF21 signaling is required for the beneficial effect of CA on hepatic TG accumulation in mice fed an HFD. We propose that FGF21 signaling potentiates the ability of CA to decrease the activation of ACC1 and HSL, key enzymes controlling the supply of long-chain fatty acid precursors for hepatic TG synthesis.
Collapse
Affiliation(s)
- Siri M Ippagunta
- Department of Biochemistry, West Virginia University, Morgantown, WV
| | | | - Andrew C Adams
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN
| | | |
Collapse
|
774
|
Meijnikman AS, Gerdes VE, Nieuwdorp M, Herrema H. Evaluating Causality of Gut Microbiota in Obesity and Diabetes in Humans. Endocr Rev 2018; 39:133-153. [PMID: 29309555 DOI: 10.1210/er.2017-00192] [Citation(s) in RCA: 192] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 12/19/2017] [Indexed: 02/06/2023]
Abstract
The pathophysiology of obesity and obesity-related diseases such as type 2 diabetes mellitus (T2DM) is complex and driven by many factors. One of the most recently identified factors in development of these metabolic pathologies is the gut microbiota. The introduction of affordable, high-throughput sequencing technologies has substantially expanded our understanding of the role of the gut microbiome in modulation of host metabolism and (cardio)metabolic disease development. Nevertheless, evidence for a role of the gut microbiome as a causal, driving factor in disease development mainly originates from studies in mouse models: data showing causality in humans are scarce. In this review, we will discuss the quality of evidence supporting a causal role for the gut microbiome in the development of obesity and diabetes, in particular T2DM, in humans. Considering overlap in potential mechanisms, the role of the gut microbiome in type 1 diabetes mellitus will also be addressed. We will elaborate on factors that drive microbiome composition in humans and discuss how alterations in microbial composition or microbial metabolite production contribute to disease development. Challenging aspects in determining causality in humans will be postulated together with strategies that might hold potential to overcome these challenges. Furthermore, we will discuss means to modify gut microbiome composition in humans to help establish causality and discuss systems biology approaches that might hold the key to unravelling the role of the gut microbiome in obesity and T2DM.
Collapse
Affiliation(s)
- Abraham S Meijnikman
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, Netherlands.,Department of Internal Medicine, MC Slotervaart, Amsterdam, Netherlands
| | - Victor E Gerdes
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, Netherlands.,Department of Internal Medicine, MC Slotervaart, Amsterdam, Netherlands
| | - Max Nieuwdorp
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, Netherlands.,Wallenberg Laboratory, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Internal Medicine, Academic Medical Center, Amsterdam, Netherlands.,Diabetes Center, Department of Internal medicine, VU University Medical Center, Amsterdam, Netherlands.,ICAR, VU University Medical Center, Amsterdam, Netherlands
| | - Hilde Herrema
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, Netherlands
| |
Collapse
|
775
|
Haluzík M, Kratochvílová H, Haluzíková D, Mráz M. Gut as an emerging organ for the treatment of diabetes: focus on mechanism of action of bariatric and endoscopic interventions. J Endocrinol 2018; 237:R1-R17. [PMID: 29378901 DOI: 10.1530/joe-17-0438] [Citation(s) in RCA: 20] [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: 12/18/2017] [Accepted: 01/29/2018] [Indexed: 01/19/2023]
Abstract
Increasing worldwide prevalence of type 2 diabetes mellitus and its accompanying pathologies such as obesity, arterial hypertension and dyslipidemia represents one of the most important challenges of current medicine. Despite intensive efforts, high percentage of patients with type 2 diabetes does not achieve treatment goals and struggle with increasing body weight and poor glucose control. While novel classes of antidiabetic medications such as incretin-based therapies and gliflozins have some favorable characteristics compared to older antidiabetics, the only therapeutic option shown to substantially modify the progression of diabetes or to achieve its remission is bariatric surgery. Its efficacy in the treatment of diabetes is well established, but the exact underlying modes of action are still only partially described. They include restriction of food amount, enhanced passage of chymus into distal part of small intestine with subsequent modification of gastrointestinal hormones and bile acids secretion, neural mechanisms, changes in gut microbiota and many other possible mechanisms underscoring the importance of the gut in the regulation of glucose metabolism. In addition to bariatric surgery, less-invasive endoscopic methods based on the principles of bariatric surgery were introduced and showed promising results. This review highlights the role of the intestine in the regulation of glucose homeostasis focusing on the mechanisms of action of bariatric and especially endoscopic methods of the treatment of diabetes. A better understanding of these mechanisms may lead to less invasive endoscopic treatments of diabetes and obesity that may complement and widen current therapeutic options.
Collapse
Affiliation(s)
- Martin Haluzík
- Centre for Experimental MedicineInstitute for Clinical and Experimental Medicine, Prague, Czech Republic
- Diabetes CentreInstitute for Clinical and Experimental Medicine, Prague, Czech Republic
- Department of Medical Biochemistry and Laboratory DiagnosticsGeneral University Hospital, Charles University in Prague, 1st Faculty of Medicine, Prague, Czech Republic
| | - Helena Kratochvílová
- Centre for Experimental MedicineInstitute for Clinical and Experimental Medicine, Prague, Czech Republic
- Department of Medical Biochemistry and Laboratory DiagnosticsGeneral University Hospital, Charles University in Prague, 1st Faculty of Medicine, Prague, Czech Republic
| | - Denisa Haluzíková
- Department of Sports MedicineGeneral University Hospital, Charles University in Prague, 1st Faculty of Medicine, Prague, Czech Republic
| | - Miloš Mráz
- Diabetes CentreInstitute for Clinical and Experimental Medicine, Prague, Czech Republic
- Department of Medical Biochemistry and Laboratory DiagnosticsGeneral University Hospital, Charles University in Prague, 1st Faculty of Medicine, Prague, Czech Republic
| |
Collapse
|
776
|
Vespasiani-Gentilucci U, Gallo P, Picardi A. The role of intestinal microbiota in the pathogenesis of NAFLD: starting points for intervention. Arch Med Sci 2018; 14:701-706. [PMID: 29765460 PMCID: PMC5949899 DOI: 10.5114/aoms.2016.58831] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 02/05/2016] [Indexed: 12/21/2022] Open
Abstract
In recent years, close links between intestinal microbiota and host metabolism have been recognized. Intestinal bacteria can participate in the extraction of calories from food, and circulation of bacterial products, in particular lipopolysaccharides (LPS), is responsible for the "metabolic endotoxemia", which contributes to insulin resistance and its complications, such as non-alcoholic fatty liver disease (NAFLD). Indeed, qualitative and quantitative intestinal dysbiotic changes have been clearly documented in NAFLD patients, and several mechanisms by which the intestinal microbiota can directly promote liver fat deposition, inflammation and fibrosis have also been described. Consistently, although with some differences concerning type and proportion of results, experimental and clinical studies are quite concordant in demonstrating beneficial effects of probiotic and/or prebiotic therapy in NAFLD. Although some physiopathological bases have been produced, major doubts still remain concerning how and when to intervene. Indeed, most of the available works were performed with mixtures of probiotics and/or prebiotics, and a baseline assessment of dysbiosis aimed at selecting the best candidates for treatment and predicting response has not been performed in any of the clinical studies in NAFLD. While future research is expected to solve these issues, the particularly favorable safety profile suggests that probiotic/prebiotic therapy could already be "tested" in NAFLD patients on an individual basis, at least once all the measures recommended by the latest guidelines have failed.
Collapse
Affiliation(s)
| | - Paolo Gallo
- Department of Medicine, Unit of Internal Medicine and Hepatology, University Campus Bio-Medico, Rome, Italy
| | - Antonio Picardi
- Department of Medicine, Unit of Internal Medicine and Hepatology, University Campus Bio-Medico, Rome, Italy
| |
Collapse
|
777
|
Kuhre RE, Wewer Albrechtsen NJ, Larsen O, Jepsen SL, Balk-Møller E, Andersen DB, Deacon CF, Schoonjans K, Reimann F, Gribble FM, Albrechtsen R, Hartmann B, Rosenkilde MM, Holst JJ. Bile acids are important direct and indirect regulators of the secretion of appetite- and metabolism-regulating hormones from the gut and pancreas. Mol Metab 2018; 11:84-95. [PMID: 29656109 PMCID: PMC6001409 DOI: 10.1016/j.molmet.2018.03.007] [Citation(s) in RCA: 156] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/03/2018] [Accepted: 03/13/2018] [Indexed: 02/06/2023] Open
Abstract
Objective Bile acids (BAs) facilitate fat absorption and may play a role in glucose and metabolism regulation, stimulating the secretion of gut hormones. The relative importance and mechanisms involved in BA-stimulated secretion of appetite and metabolism regulating hormones from the gut and pancreas is not well described and was the purpose of this study. Methods The effects of bile acids on the secretion of gut and pancreatic hormones was studied in rats and compared to the most well described nutritional secretagogue: glucose. The molecular mechanisms that underlie the secretion was studied by isolated perfused rat and mouse small intestine and pancreas preparations and supported by immunohistochemistry, expression analysis, and pharmacological studies. Results Bile acids robustly stimulate secretion of not only the incretin hormones, glucose-dependent insulinotropic peptide (GIP), and glucagon-like peptide-1 (GLP-1), but also glucagon and insulin in vivo, to levels comparable to those resulting from glucose stimulation. The mechanisms of GLP-1, neurotensin, and peptide YY (PYY) secretion was secondary to intestinal absorption and depended on activation of basolateral membrane Takeda G-protein receptor 5 (TGR5) receptors on the L-cells in the following order of potency: Lithocholic acid (LCA) >Deoxycholicacid (DCA)>Chenodeoxycholicacid (CDCA)> Cholic acid (CA). Thus BAs did not stimulate secretion of GLP-1 and PYY from perfused small intestine in TGR5 KO mice but stimulated robust responses in wild type littermates. TGR5 is not expressed on α-cells or β-cells, and BAs had no direct effects on glucagon or insulin secretion from the perfused pancreas. Conclusion BAs should be considered not only as fat emulsifiers but also as important regulators of appetite- and metabolism-regulating hormones by activation of basolateral intestinal TGR5. Bile acids stimulate the secretion of metabolism-regulating hormones from the gut. Bile acids stimulate secretion of gut hormones to a similar extent as glucose. Activation of basolateral TGR5 receptors mediates the responses. Bile acids stimulate glucagon and insulin secretion, but only indirectly. Bile acids should be regarded as important regulators of blood glucose and metabolism.
Collapse
Affiliation(s)
- Rune E Kuhre
- Department of Biomedical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark; NNF Center for Basic Metabolic Research, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Nicolai J Wewer Albrechtsen
- Department of Biomedical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark; NNF Center for Basic Metabolic Research, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Olav Larsen
- Department of Biomedical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Sara L Jepsen
- Department of Biomedical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark; NNF Center for Basic Metabolic Research, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Emilie Balk-Møller
- Department of Biomedical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark; NNF Center for Basic Metabolic Research, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Daniel B Andersen
- Department of Biomedical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark; NNF Center for Basic Metabolic Research, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Carolyn F Deacon
- Department of Biomedical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark; NNF Center for Basic Metabolic Research, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Kristina Schoonjans
- Laboratory of Metabolic Signaling, Ecole Polytechnique Fédérale de Lausanne, Station 15, CH-1015, Lausanne, Switzerland
| | - Frank Reimann
- Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Wellcome Trust-Medical Research Council, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, CB2 0QQ, United Kingdom
| | - Fiona M Gribble
- Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Wellcome Trust-Medical Research Council, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, CB2 0QQ, United Kingdom
| | - Reidar Albrechtsen
- Department of Biomedical Sciences, and Biotech Research and Innovation Centre (BRIC), University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Bolette Hartmann
- Department of Biomedical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark; NNF Center for Basic Metabolic Research, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Mette M Rosenkilde
- Department of Biomedical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark; NNF Center for Basic Metabolic Research, University of Copenhagen, DK-2200, Copenhagen, Denmark.
| |
Collapse
|
778
|
Chu H, Williams B, Schnabl B. Gut microbiota, fatty liver disease, and hepatocellular carcinoma. LIVER RESEARCH 2018; 2:43-51. [PMID: 30416839 PMCID: PMC6223644 DOI: 10.1016/j.livres.2017.11.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Intestinal bacteria contribute to the pathogenesis of non-alcoholic fatty liver disease (NAFLD). Recently developed microbial profiling techniques are beginning to shed light on the nature of the changes in the gut microbiota that accompany NAFLD and non-alcoholic steatohepatitis (NASH). In this review, we summarize the role of gut microbiota in the development of NAFLD, NASH, and hepatocellular carcinoma (HCC). We highlight the mechanisms by which gut microbiota contribute to NAFLD/NASH, including through alterations in gut epithelial permeability, choline metabolism, endogenous alcohol production, release of inflammatory cytokines, regulation of hepatic Toll-like receptor (TLR), and bile acid metabolism. In addition, we analyze possible mechanisms for enhanced hepatic carcinogenesis, including alterations in bile acid metabolism, release of inflammatory cytokines, and expression of TLR-4. Finally, we describe therapeutic approaches for NAFLD/NASH and preventive strategies for HCC involving modulation of the intestinal microbiota or affected host pathways. Although recent studies have provided useful information, large-scale prospective studies are required to better characterize the intestinal microbiota and metabolome, in order to demonstrate a causative role for changes in the gut microbiota in the etiology of NAFLD/NASH, to identify new therapeutic strategies for NAFLD/NASH, and to develop more effective methods of preventing HCC.
Collapse
Affiliation(s)
- Huikuan Chu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Brandon Williams
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA, USA,epartment of Medicine, VA San Diego Healthcare System, San Diego, CA, USA,Corresponding author. Department of Medicine, University of California San Diego, Biomedical Research Facility 2 (BRF2), La Jolla, CA, USA. (B. Schnabl)
| |
Collapse
|
779
|
Kumar S, Palaia T, Hall C, Ragolia L. DP1 receptor agonist, BW245C inhibits diet-induced obesity in ApoE -/- mice. Obes Res Clin Pract 2018; 12:229-241. [PMID: 28602634 DOI: 10.1016/j.orcp.2017.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 05/11/2017] [Accepted: 05/12/2017] [Indexed: 01/22/2023]
Abstract
BACKGROUND/OBJECTIVE Lipocalin Prostaglandin D2 synthase (LPGDS) contributes to the production of PGD2, which has been associated with adipogenesis. In this study, we aimed to investigate the role of PGD2 on obesity through its DP1 and DP2 receptor signaling using intraperitoneal injection of their respective agonists and antagonists. METHODS ApoE-/- mice were divided into five groups: vehicle control (n=5), DP1 receptor agonist (n=5), DP1 receptor antagonist (n=5), DP2 receptor agonist (n=5), and DP2 receptor antagonist (n=5), and the study was carried out for 10 weeks. RESULTS Despite being on high fat diet, mice receiving DP1 receptor agonist sustained a significant inhibition of weight gain throughout the study gaining only 11.4% body weight compared to the controls gaining 61% body weight. Interestingly, parallel to the body weight, the DP1 receptor agonist group showed a significant reduction in food intake throughout the study. Consistently, fasting leptin, insulin and bile acids levels were elevated in the DP1 receptor agonist group compared to controls. As expected, there was a significant reduction in fasting glucose level in DP1 receptor agonist group. At last, as a result of weight gain inhibition, DP1 receptor agonist also imparted cardiovascular benefits showing significant reduction in aortic wall thickness, intima, adventia and lumen size. CONCLUSION Based on the obtained results, we believe DP1 receptor agonism inhibited diet induced weight gain possibly through controlling appetite which consequently imparted beneficial cardiometabolic effects. DP1 receptor agonism may represent a novel therapeutic target for the management of obesity.
Collapse
Affiliation(s)
- Sunil Kumar
- Department of Biomedical Research, Winthrop University Hospital, Mineola, NY 11501, United States
| | - Thomas Palaia
- Department of Biomedical Research, Winthrop University Hospital, Mineola, NY 11501, United States
| | - Christopher Hall
- Department of Biomedical Research, Winthrop University Hospital, Mineola, NY 11501, United States
| | - Louis Ragolia
- Department of Biomedical Research, Winthrop University Hospital, Mineola, NY 11501, United States; Department of Medicine, Stony Brook University School of Medicine, Stony Brook, NY 11794, United States.
| |
Collapse
|
780
|
Nonalcoholic fatty liver disease, cholesterol gallstones, and cholecystectomy: new insights on a complex relationship. Curr Opin Gastroenterol 2018; 34:90-96. [PMID: 29266009 DOI: 10.1097/mog.0000000000000416] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Gallstone disease (GSD) and nonalcoholic fatty liver disease (NAFLD often coexist in a given patient and both conditions are associated to obesity and insulin resistance. The relationship between GSD and NAFLD is complex and bidirectional. In the present review, we summarize the existing information on the complex link between GSD and NAFLD and the potential implications for patient care. RECENT FINDINGS Several clinical studies and systematic reviews have addressed the association between NAFLD and GSD underscoring that NAFLD is an independent risk factor for GSD. Conversely, GSD has been found also to be an independent risk factor for NAFLD with GSD potentially being linked to greater disease severity. In addition to the data showing association of NAFLD and GSD, recent evidence has also showed that cholecystectomy may itself be a risk factor for NAFLD development. The complex and bidirectional relationship between these diseases is partially explained by a number of common pathogenic links but the precise underlying mechanisms of the association of GSD and NAFLD need to be better delineated. Also, although the mechanisms of the promotional effect of cholecystectomy on NAFLD development are unknown, recent findings unveiling new aspects of gallbladder physiology and endocrine actions of bile acids provide a framework to advance research in this field. SUMMARY In this review, we address the different aspects of the complex association between NAFLD and GSD. The potential underlying mechanisms and recent information on endocrine actions of bile acids and the gallbladder are reviewed.
Collapse
|
781
|
Chevre R, Trigueros-Motos L, Castaño D, Chua T, Corlianò M, Patankar JV, Sng L, Sim L, Juin TL, Carissimo G, Ng LFP, Yi CNJ, Eliathamby CC, Groen AK, Hayden MR, Singaraja RR. Therapeutic modulation of the bile acid pool by Cyp8b1 knockdown protects against nonalcoholic fatty liver disease in mice. FASEB J 2018; 32:3792-3802. [PMID: 29481310 DOI: 10.1096/fj.201701084rr] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Bile acids (BAs) are surfactant molecules that regulate the intestinal absorption of lipids. Thus, the modulation of BAs represents a potential therapy for nonalcoholic fatty liver disease (NAFLD), which is characterized by hepatic accumulation of fat and is a major cause of liver disease worldwide. Cyp8b1 is a critical modulator of the hydrophobicity index of the BA pool. As a therapeutic proof of concept, we aimed to determine the impact of Cyp8b1 inhibition in vivo on BA pool composition and as protection against NAFLD. Inhibition of Cyp8b1 expression in mice led to a remodeling of the BA pool, which altered its signaling properties and decreased intestinal fat absorption. In a model of cholesterol-induced NAFLD, Cyp8b1 knockdown significantly decreased steatosis and hepatic lipid content, which has been associated with an increase in fecal lipid and BA excretion. Moreover, inhibition of Cyp8b1 not only decreased hepatic lipid accumulation, but also resulted in the clearance of previously accumulated hepatic cholesterol, which led to a regression in hepatic steatosis. Taken together, our data demonstrate that Cyp8b1 inhibition is a viable therapeutic target of crucial interest for metabolic diseases, such as NAFLD.-Chevre, R., Trigueros-Motos, L., Castaño, D., Chua, T., Corlianò, M., Patankar, J. V., Sng, L., Sim, L., Juin, T. L., Carissimo, G., Ng, L. F. P., Yi, C. N. J., Eliathamby, C. C., Groen, A. K., Hayden, M. R., Singaraja, R. R. Therapeutic modulation of the bile acid pool by Cyp8b1 knockdown protects against nonalcoholic fatty liver disease in mice.
Collapse
Affiliation(s)
- Raphael Chevre
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology, and Research (A*STAR), Singapore
| | - Laia Trigueros-Motos
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology, and Research (A*STAR), Singapore
| | - David Castaño
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology, and Research (A*STAR), Singapore
| | - Tricia Chua
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology, and Research (A*STAR), Singapore
| | - Maria Corlianò
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology, and Research (A*STAR), Singapore
| | - Jay V Patankar
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lareina Sng
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology, and Research (A*STAR), Singapore
| | - Lauren Sim
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology, and Research (A*STAR), Singapore
| | - Tan Liang Juin
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology, and Research (A*STAR), Singapore
| | - Guillaume Carissimo
- Singapore Immunology Network, Agency for Science, Technology, and Research (A*STAR), Singapore
| | - Lisa F P Ng
- Singapore Immunology Network, Agency for Science, Technology, and Research (A*STAR), Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Cheryl Neo Jia Yi
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology, and Research (A*STAR), Singapore
| | - Chelsea Chandani Eliathamby
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology, and Research (A*STAR), Singapore
| | - Albert K Groen
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; and.,Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Michael R Hayden
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology, and Research (A*STAR), Singapore.,Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Roshni R Singaraja
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology, and Research (A*STAR), Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
782
|
Affiliation(s)
- Leszek Szablewski
- Medical University of Warsaw, Department of General Biology and Parasitology, Warsaw, Poland
| |
Collapse
|
783
|
Weng SG, Zhang B, Wang X, Chen H. Effects of Duodenal-Jejunal Exclusion and New Bilio-Pancreatic Diversion on Blood Glucose in Rats with Type 2 Diabetes Mellitus. Obes Surg 2018; 27:2067-2072. [PMID: 28233263 DOI: 10.1007/s11695-017-2599-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVE The current study aimed to investigate the effects of duodenal-jejunal bypass (DJB), new bilio-pancreatic diversion (NBPD), and duodenal-jejunal exclusion (DJE) on blood glucose in rats with type 2 diabetes mellitus (T2DM). METHODS Male Sprague Dawley rats were fed with high glucose, high fat food, and intraperitoneally injected with streptozotocin to establish a T2DM animal model. T2DM rats were randomly assigned into 4 groups: a sham group (n = 8), DJB group (n = 9), NBPD group (n = 10), and DJE group (n = 10). Body weight, 2-h postprandial glucose, oral glucose tolerance, fasting serum bile acid, 2-h postprandial serum bile acid, fasting insulin, 2-h postprandial insulin (INS), fasting glucagon-like peptide-1 (GLP-1), and 2-h postprandial GLP-1 were measured before and after surgery. RESULTS Six weeks after surgery, the 2-h postprandial glucose in the DJB (16.1 ± 6.7 mmol/L) and NBPD (19.5 ± 5.7 mmol/L) groups decreased significantly compared to the sham group (25.8 ± 4.9 mmol/L) (P < 0.05). There was no significant difference between the DJE (25.0 ± 5.0 mmol/L) and sham groups (P > 0.05). Four weeks after surgery, fasting serum bile acid in the DJB group (60.6 ± 11.4 μmol/L) and NBPD group (54.4 ± 7.64 μmol/L) was significantly higher than that in the sham group (34.3 ± 6.98 μmol/L; P < 0.05). However, fasting GLP-1, 2-h postprandial GLP-1, and insulin remained unchanged at different time points after surgery (P > 0.05). Body weight remained stable after surgery in all 4 groups (P > 0.05). CONCLUSION NBPD plays a major role in the therapy of T2DM with DJB. NBPD may significantly increase fasting serum bile acid in T2DM rats, an action that may be one of the mechanisms underlying the therapeutic effects of DJB on T2DM.
Collapse
Affiliation(s)
- Shan Geng Weng
- Hepatopancreatobiliary Surgery Department, the First Affiliated Hospital of Fujian Medical University, No 20 Chazhong Road, Fuzhou City, Fujian, People's Republic of China.
| | - Bin Zhang
- Hepatopancreatobiliary Surgery Department, the First Affiliated Hospital of Fujian Medical University, No 20 Chazhong Road, Fuzhou City, Fujian, People's Republic of China
| | - Xiaojian Wang
- Hepatopancreatobiliary Surgery Department, the First Affiliated Hospital of Fujian Medical University, No 20 Chazhong Road, Fuzhou City, Fujian, People's Republic of China
| | - Hao Chen
- Hepatopancreatobiliary Surgery Department, the First Affiliated Hospital of Fujian Medical University, No 20 Chazhong Road, Fuzhou City, Fujian, People's Republic of China
| |
Collapse
|
784
|
Teno N, Yamashita Y, Iguchi Y, Fujimori K, Une M, Nishimaki-Mogami T, Hiramoto T, Gohda K. Nonacidic Chemotype Possessing N-Acylated Piperidine Moiety as Potent Farnesoid X Receptor (FXR) Antagonists. ACS Med Chem Lett 2018; 9:78-83. [PMID: 29456791 DOI: 10.1021/acsmedchemlett.7b00363] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Accepted: 12/27/2017] [Indexed: 12/20/2022] Open
Abstract
Farnesoid X receptor (FXR) plays a major role in the control of cholesterol metabolism. Antagonizing transcriptional activity of FXR is an effective means to treat the relevant metabolic syndrome. Some of antagonists so far have the charged functions; however, they may negatively affect the pharmacokinetics. We describe herein a structure-activity relationship (SAR) exploration of nonacidic FXR antagonist 6 focusing on two regions in the structure and biological evaluation of nonacidic 10 with the characteristic N-acylated piperidine group obtained from SAR studies. As the robust affinity to FXR is feasible with our nonacidic analogue, 10 is among the most promising candidates for in vivo testing.
Collapse
Affiliation(s)
| | | | | | - Ko Fujimori
- Faculty
of Pharmaceutical Sciences, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka 569-1094, Japan
| | | | | | | | - Keigo Gohda
- Computer-aided
Molecular Modeling Research Center, Kansai (CAMM-Kansai), 3-32-302
Tsuto, Otsuka, Nishinomiya 663-8241, Japan
| |
Collapse
|
785
|
Lu Y, Zheng W, Lin S, Guo F, Zhu Y, Wei Y, Liu X, Jin S, Jin L, Li Y. Identification of an Oleanane-Type Triterpene Hedragonic Acid as a Novel Farnesoid X Receptor Ligand with Liver Protective Effects and Anti-inflammatory Activity. Mol Pharmacol 2018; 93:63-72. [PMID: 29162643 DOI: 10.1124/mol.117.109900] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 11/15/2017] [Indexed: 02/14/2025] Open
Abstract
Farnesoid X receptor (FXR) and G-protein-coupled bile acid receptor 1 (GPBAR1) are two important bile acid (BA) receptors. As non-BAs drug template for GPBAR1, none of the natural oleanane-type triterpenes have been reported as FXR ligands, despite FXR and GPBAR1 having similar binding pockets for BAs. Here, we report the natural triterpene hedragonic acid that has been isolated from the stem and root of Celastrus orbiculatus Thunb. (COT) as an effective agonist for FXR. Both biochemical amplified luminescent proximity homogeneous assay and cell-based reporter assays showed that hedragonic acid regulated the transcriptional activity of FXR. Circular dichroism spectroscopy further suggested the conformational changes of FXR upon the binding of hedragonic acid. Interestingly, the crystal structure of hedragonic acid-bound FXR revealed a unique binding mode with hedragonic acid occupying a novel binding pocket different from the classic binding position. The structural comparison between hedragonic acid-bound FXR and oleanolic acid-bound GPBAR1 explained the molecular basis for the selectivity of oleanane-type triterpenes for FXR. Moreover, hedragonic acid treatment protected mice from liver injury induced by acetaminophen overdose and decreased hepatic inflammatory responses in an FXR-dependent manner, suggesting that hedragonic acid might be one of the major components of COT for its multifunctional pharmaceutical uses. In conclusion, our results provide novel structure templates for drug design based on natural triterpenes by targeting FXR and/or GPBAR1 with pharmaceutical values.
Collapse
Affiliation(s)
- Yi Lu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Innovation Center for Cell Signaling Network, Xiamen University, Xiamen, Fujian, China
| | - Weili Zheng
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Innovation Center for Cell Signaling Network, Xiamen University, Xiamen, Fujian, China
| | - Shengchen Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Innovation Center for Cell Signaling Network, Xiamen University, Xiamen, Fujian, China
| | - Fusheng Guo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Innovation Center for Cell Signaling Network, Xiamen University, Xiamen, Fujian, China
| | - Yanlin Zhu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Innovation Center for Cell Signaling Network, Xiamen University, Xiamen, Fujian, China
| | - Yijuan Wei
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Innovation Center for Cell Signaling Network, Xiamen University, Xiamen, Fujian, China
| | - Xi Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Innovation Center for Cell Signaling Network, Xiamen University, Xiamen, Fujian, China
| | - Shikai Jin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Innovation Center for Cell Signaling Network, Xiamen University, Xiamen, Fujian, China
| | - Lihua Jin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Innovation Center for Cell Signaling Network, Xiamen University, Xiamen, Fujian, China
| | - Yong Li
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Innovation Center for Cell Signaling Network, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
786
|
Puri P, Daita K, Joyce A, Mirshahi F, Santhekadur PK, Cazanave S, Luketic VA, Siddiqui MS, Boyett S, Min HK, Kumar DP, Kohli R, Zhou H, Hylemon PB, Contos MJ, Idowu M, Sanyal AJ. The presence and severity of nonalcoholic steatohepatitis is associated with specific changes in circulating bile acids. Hepatology 2018; 67:534-548. [PMID: 28696585 PMCID: PMC5764808 DOI: 10.1002/hep.29359] [Citation(s) in RCA: 299] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 05/25/2017] [Accepted: 07/07/2017] [Indexed: 12/12/2022]
Abstract
The histologic spectrum of nonalcoholic fatty liver disease (NAFLD) includes fatty liver (NAFL) and steatohepatitis (NASH), which can progress to cirrhosis in up to 20% of NASH patients. Bile acids (BA) are linked to the pathogenesis and therapy of NASH. We (1) characterized the plasma BA profile in biopsy-proven NAFL and NASH and compared to controls and (2) related the plasma BA profile to liver histologic features, disease activity, and fibrosis. Liquid chromatography/mass spectrometry quantified BAs. Descriptive statistics, paired and multiple group comparisons, and regression analyses were performed. Of 86 patients (24 controls, 25 NAFL, and 37 NASH; mean age 51.8 years and body mass index 31.9 kg/m2 ), 66% were women. Increased total primary BAs and decreased secondary BAs (both P < 0.05) characterized NASH. Total conjugated primary BAs were significantly higher in NASH versus NAFL (P = 0.047) and versus controls (P < 0.0001). NASH had higher conjugated to unconjugated chenodeoxycholate (P = 0.04), cholate (P = 0.0004), and total primary BAs (P < 0.0001). The total cholate to chenodeoxycholate ratio was significantly higher in NAFLD without (P = 0.005) and with (P = 0.02) diabetes. Increased key BAs were associated with higher grades of steatosis (taurocholate), lobular (glycocholate) and portal inflammation (taurolithocholate), and hepatocyte ballooning (taurocholate). Conjugated cholate and taurocholate directly and secondary to primary BA ratio inversely correlated to NAFLD activity score. A higher ratio of total secondary to primary BA decreased (odds ratio, 0.57; P = 0.004) and higher conjugated cholate increased the likelihood of significant fibrosis (F≥2) (P = 0.007). Conclusion: NAFLD is associated with significantly altered circulating BA composition, likely unaffected by type 2 diabetes, and correlated with histological features of NASH; these observations provide the foundation for future hypothesis-driven studies of specific effects of BAs on specific aspects of NASH. (Hepatology 2018;67:534-548).
Collapse
Affiliation(s)
- Puneet Puri
- Div. of Gastroenterology, Hepatology and Nutrition, Dept. of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Kalyani Daita
- Div. of Gastroenterology, Hepatology and Nutrition, Dept. of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | | | - Faridoddin Mirshahi
- Div. of Gastroenterology, Hepatology and Nutrition, Dept. of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Prasanna K. Santhekadur
- Div. of Gastroenterology, Hepatology and Nutrition, Dept. of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Sophie Cazanave
- Div. of Gastroenterology, Hepatology and Nutrition, Dept. of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Velimir A Luketic
- Div. of Gastroenterology, Hepatology and Nutrition, Dept. of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Mohammad S. Siddiqui
- Div. of Gastroenterology, Hepatology and Nutrition, Dept. of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Sherry Boyett
- Div. of Gastroenterology, Hepatology and Nutrition, Dept. of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Hae-Ki Min
- Div. of Gastroenterology, Hepatology and Nutrition, Dept. of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Divya P. Kumar
- Div. of Gastroenterology, Hepatology and Nutrition, Dept. of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Rohit Kohli
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Children’s Hospital Los Angeles, University of Southern California, Los Angeles, CA
| | - Huiping Zhou
- Div. of Gastroenterology, Hepatology and Nutrition, Dept. of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Phillip B. Hylemon
- Div. of Gastroenterology, Hepatology and Nutrition, Dept. of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Melissa J Contos
- Div. of Gastroenterology, Hepatology and Nutrition, Dept. of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Michael Idowu
- Div. of Gastroenterology, Hepatology and Nutrition, Dept. of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Arun J. Sanyal
- Div. of Gastroenterology, Hepatology and Nutrition, Dept. of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| |
Collapse
|
787
|
Leong KSW, Derraik JGB, Hofman PL, Cutfield WS. Antibiotics, gut microbiome and obesity. Clin Endocrinol (Oxf) 2018; 88:185-200. [PMID: 29023853 DOI: 10.1111/cen.13495] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 10/09/2017] [Indexed: 12/13/2022]
Abstract
Antibiotics have been hailed by many as "miracle drugs" that have been effectively treating infectious diseases for over a century, leading to a marked reduction in morbidity and mortality. However, with the increasing use of antibiotics, we are now faced not only with the increasing threat of antibiotic resistance, but also with a rising concern about potential long-term effects of antibiotics on human health, including the development of obesity. The obesity pandemic continues to increase, a problem that affects both adults and children alike. Disruptions to the gut microbiome have been linked to a multitude of adverse conditions, including obesity, type 2 diabetes, inflammatory bowel diseases, anxiety, autism, allergies, and autoimmune diseases. This review focuses on the association between antibiotics and obesity, and the role of the gut microbiome. There is strong evidence supporting the role of antibiotics in the development of obesity in well-controlled animal models. However, evidence for this link in humans is still inconclusive, and we need further well-designed clinical trials to clarify this association.
Collapse
Affiliation(s)
- Karen S W Leong
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - José G B Derraik
- Liggins Institute, University of Auckland, Auckland, New Zealand
- A Better Start - National Science Challenge, University of Auckland, Auckland, New Zealand
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Paul L Hofman
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Wayne S Cutfield
- Liggins Institute, University of Auckland, Auckland, New Zealand
- A Better Start - National Science Challenge, University of Auckland, Auckland, New Zealand
| |
Collapse
|
788
|
Eggink HM, Tambyrajah LL, van den Berg R, Mol IM, van den Heuvel JK, Koehorst M, Groen AK, Boelen A, Kalsbeek A, Romijn JA, Rensen PCN, Kooijman S, Soeters MR. Chronic infusion of taurolithocholate into the brain increases fat oxidation in mice. J Endocrinol 2018; 236:85-97. [PMID: 29233934 DOI: 10.1530/joe-17-0503] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 12/11/2017] [Indexed: 01/11/2023]
Abstract
Bile acids can function in the postprandial state as circulating signaling molecules in the regulation of glucose and lipid metabolism via the transmembrane receptor TGR5 and nuclear receptor FXR. Both receptors are present in the central nervous system, but their function in the brain is unclear. Therefore, we investigated the effects of intracerebroventricular (i.c.v.) administration of taurolithocholate (tLCA), a strong TGR5 agonist, and GW4064, a synthetic FXR agonist, on energy metabolism. We determined the effects of chronic i.c.v. infusion of tLCA, GW4064, or vehicle on energy expenditure, body weight and composition as well as tissue specific fatty acid uptake in mice equipped with osmotic minipumps. We found that i.c.v. administration of tLCA (final concentration in cerebrospinal fluid: 1 μM) increased fat oxidation (tLCA group: 0.083 ± 0.006 vs control group: 0.036 ± 0.023 kcal/h, F = 5.46, P = 0.04) and decreased fat mass (after 9 days of tLCA infusion: 1.35 ± 0.13 vs controls: 1.96 ± 0.23 g, P = 0.03). These changes were associated with enhanced uptake of triglyceride-derived fatty acids by brown adipose tissue and with browning of subcutaneous white adipose tissue. I.c.v. administration of GW4064 (final concentration in cerebrospinal fluid: 10 μM) did not affect energy metabolism, body composition nor bile acid levels, negating a role of FXR in the central nervous system in metabolic control. In conclusion, bile acids such as tLCA may exert metabolic effects on fat metabolism via the brain.
Collapse
Affiliation(s)
- Hannah M Eggink
- Department of Endocrinology and MetabolismAcademic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
- Hypothalamic Integration MechanismsNetherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Lauren L Tambyrajah
- Division of EndocrinologyDepartment of Medicine, Leiden University Medical Centre, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical Centre, Leiden, The Netherlands
| | - Rosa van den Berg
- Division of EndocrinologyDepartment of Medicine, Leiden University Medical Centre, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical Centre, Leiden, The Netherlands
| | - Isabel M Mol
- Division of EndocrinologyDepartment of Medicine, Leiden University Medical Centre, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical Centre, Leiden, The Netherlands
| | - Jose K van den Heuvel
- Division of EndocrinologyDepartment of Medicine, Leiden University Medical Centre, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical Centre, Leiden, The Netherlands
| | - Martijn Koehorst
- Department of Pediatrics and Laboratory MedicineUniversity Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Albert K Groen
- Department of Pediatrics and Laboratory MedicineUniversity Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
- Department of Vascular MedicineAmsterdam Diabetes Centre, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Anita Boelen
- Department of Endocrinology and MetabolismAcademic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and MetabolismAcademic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
- Hypothalamic Integration MechanismsNetherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Johannes A Romijn
- Department of MedicineAcademic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Patrick C N Rensen
- Division of EndocrinologyDepartment of Medicine, Leiden University Medical Centre, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical Centre, Leiden, The Netherlands
| | - Sander Kooijman
- Division of EndocrinologyDepartment of Medicine, Leiden University Medical Centre, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical Centre, Leiden, The Netherlands
| | - Maarten R Soeters
- Department of Endocrinology and MetabolismAcademic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
789
|
Brønden A, Albér A, Rohde U, Gasbjerg LS, Rehfeld JF, Holst JJ, Vilsbøll T, Knop FK. The bile acid-sequestering resin sevelamer eliminates the acute GLP-1 stimulatory effect of endogenously released bile acids in patients with type 2 diabetes. Diabetes Obes Metab 2018; 20:362-369. [PMID: 28786523 DOI: 10.1111/dom.13080] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 08/01/2017] [Accepted: 08/02/2017] [Indexed: 12/27/2022]
Abstract
AIMS Discovery of the specific bile acid receptors farnesoid X receptor (FXR) and Takeda G protein-coupled receptor 5 (TGR5) in enteroendocrine L cells has prompted research focusing on the impact of bile acids on glucagon-like peptide-1 (GLP-1) secretion and glucose metabolism. The aim of the present study was to assess the GLP-1 secretory and gluco-metabolic effects of endogenously released bile, with and without concomitant administration of the bile acid-sequestering resin, sevelamer, in patients with type 2 diabetes. MATERIALS AND METHODS We performed a randomized, placebo-controlled, double-blinded cross-over study including 15 metformin-treated patients with type 2 diabetes. During 4 experimental study days, either sevelamer 3200 mg or placebo in combination with intravenous infusion of cholecystokinin (CCK) (0.4 pmol sulfated CCK-8/kg/min) or saline was administered in randomized order. The primary endpoint was plasma GLP-1 excursions as measured by incremental area under the curve. Secondary endpoints included plasma responses of glucose, triglycerides, insulin, CCK, fibroblast growth factor-19 and 7α-hydroxy-4-cholesten-3-one (C4). In addition, gallbladder dynamics, gastric emptying, resting energy expenditure, appetite and ad libitum food intake were assessed. RESULTS CCK-mediated gallbladder emptying was demonstrated to elicit a significant induction of GLP-1 secretion compared to saline, whereas concomitant single-dose administration of the bile acid sequestrant sevelamer was shown to eliminate the acute bile acid-induced increase in plasma GLP-1 excursions. CONCLUSIONS Single-dose administration of sevelamer eliminated bile acid-mediated GLP-1 secretion in patients with type 2 diabetes, which could be explained by reduced bile acid stimulation of the basolaterally localized TGR5 on enteroendocrine L cells.
Collapse
Affiliation(s)
- Andreas Brønden
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Anders Albér
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Ulrich Rohde
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Laerke S Gasbjerg
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens F Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tina Vilsbøll
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, University of Copenhagen, Copenhagen, Denmark
| | - Filip K Knop
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
790
|
Abstract
Nonalcoholic fatty liver disease is the most common cause of chronic liver disease in North America and is growing as a cause of chronic liver disease in many other parts of the world as well. It has 2 principal clinical-pathologic phenotypes: (1) nonalcoholic fatty liver and (2) nonalcoholic steatohepatitis. The development of both phenotypes is tightly linked to excess body weight and insulin resistance. This review discusses the emerging tools for the analysis of the microbiome, their limitations, and the existing literature with respect to the intestinal microbiome and their role in nonalcoholic fatty liver.
Collapse
|
791
|
Luo P, Yin P, Hua R, Tan Y, Li Z, Qiu G, Yin Z, Xie X, Wang X, Chen W, Zhou L, Wang X, Li Y, Chen H, Gao L, Lu X, Wu T, Wang H, Niu J, Xu G. A Large-scale, multicenter serum metabolite biomarker identification study for the early detection of hepatocellular carcinoma. Hepatology 2018; 67:662-675. [PMID: 28960374 PMCID: PMC6680350 DOI: 10.1002/hep.29561] [Citation(s) in RCA: 281] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 08/29/2017] [Accepted: 09/24/2017] [Indexed: 12/13/2022]
Abstract
Hepatocellular carcinoma (HCC) is the third most lethal cancer worldwide. The lack of effective biomarkers for the early detection of HCC results in unsatisfactory curative treatments. Here, metabolite biomarkers were identified and validated for HCC diagnosis. A total of 1,448 subjects, including healthy controls and patients with chronic hepatitis B virus infection, liver cirrhosis, and HCC, were recruited from multiple centers in China. Liquid chromatography-mass spectrometry-based metabolomics methods were used to characterize the subjects' serum metabolic profiles and to screen and validate the HCC biomarkers. A serum metabolite biomarker panel including phenylalanyl-tryptophan and glycocholate was defined. This panel had a higher diagnostic performance than did α-fetoprotein (AFP) in differentiating HCC from a high-risk population of cirrhosis, such as an area under the receiver-operating characteristic curve of 0.930, 0.892, and 0.807 for the panel versus 0.657, 0.725, and 0.650 for AFP in the discovery set, test set, and cohort 1 of the validation set, respectively. In the nested case-control study, this panel had high sensitivity (range 80.0%-70.3%) to detect preclinical HCC, and its combination with AFP provided better risk prediction of preclinical HCC before clinical diagnosis. Besides, this panel showed a larger area under the receiver-operating characteristic curve than did AFP (0.866 versus 0.682) to distinguish small HCC, and 80.6% of the AFP false-negative patients with HCC were correctly diagnosed using this panel in the test set, which was corroborated by the validation set. The specificity and biological relevance of the identified biomarkers were further evaluated using sera from another two cancers and HCC tissue specimens, respectively. Conclusion: The discovered and validated serum metabolite biomarker panel exhibits good diagnostic performance for the early detection of HCC from at-risk populations. (Hepatology 2018;67:662-675).
Collapse
Affiliation(s)
- Ping Luo
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina,University of Chinese Academy of SciencesBeijingChina
| | - Peiyuan Yin
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
| | - Rui Hua
- Department of Hepatology, First HospitalJilin UniversityChangchunJilinChina
| | - Yexiong Tan
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery InstituteThe Second Military Medical UniversityShanghaiChina
| | - Zaifang Li
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina,University of Chinese Academy of SciencesBeijingChina
| | - Gaokun Qiu
- MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical CollegeHuazhong University of Science & TechnologyWuhanHubeiChina
| | - Zhenyu Yin
- Zhongshan Hospital of Xiamen UniversityXiamenChina
| | | | - Xiaomei Wang
- Department of Hepatology, First HospitalJilin UniversityChangchunJilinChina
| | - Wenbin Chen
- Shangdong Provincial Hospital Affiliated to Shandong UniversityJinanChina
| | - Lina Zhou
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
| | - Xiaolin Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
| | - Yanli Li
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
| | | | - Ling Gao
- Shangdong Provincial Hospital Affiliated to Shandong UniversityJinanChina
| | - Xin Lu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
| | - Tangchun Wu
- MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical CollegeHuazhong University of Science & TechnologyWuhanHubeiChina
| | - Hongyang Wang
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery InstituteThe Second Military Medical UniversityShanghaiChina
| | - Junqi Niu
- Department of Hepatology, First HospitalJilin UniversityChangchunJilinChina
| | - Guowang Xu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
| |
Collapse
|
792
|
Marra F, Svegliati-Baroni G. Lipotoxicity and the gut-liver axis in NASH pathogenesis. J Hepatol 2018; 68:280-295. [PMID: 29154964 DOI: 10.1016/j.jhep.2017.11.014] [Citation(s) in RCA: 598] [Impact Index Per Article: 85.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 11/05/2017] [Accepted: 11/09/2017] [Indexed: 12/12/2022]
Abstract
The pathogenesis of non-alcoholic fatty liver disease, particularly the mechanisms whereby a minority of patients develop a more severe phenotype characterised by hepatocellular damage, inflammation, and fibrosis is still incompletely understood. Herein, we discuss two pivotal aspects of the pathogenesis of NASH. We first analyse the initial mechanisms responsible for hepatocellular damage and inflammation, which derive from the toxic effects of excess lipids. Accumulating data indicate that the total amount of triglycerides stored in hepatocytes is not the major determinant of lipotoxicity, and that specific lipid classes act as damaging agents on liver cells. In particular, the role of free fatty acids such as palmitic acid, cholesterol, lysophosphatidylcholine and ceramides has recently emerged. These lipotoxic agents affect the cell behaviour via multiple mechanisms, including activation of signalling cascades and death receptors, endoplasmic reticulum stress, modification of mitochondrial function, and oxidative stress. In the second part of this review, the cellular and molecular players involved in the cross-talk between the gut and the liver are considered. These include modifications to the microbiota, which provide signals through the intestine and bacterial products, as well as hormones produced in the bowel that affect metabolism at different levels including the liver. Finally, the activation of nuclear receptors by bile acids is analysed.
Collapse
Affiliation(s)
- Fabio Marra
- Dipartimento di Medicina Sperimentale e Clinica and Centro di Ricerca Denothe, Università di Firenze, Italy.
| | - Gianluca Svegliati-Baroni
- Dipartimento Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Ancona, Italy; Centro Interdipartimentale Obesità, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
793
|
Issa D, Patel V, Sanyal AJ. Future therapy for non-alcoholic fatty liver disease. Liver Int 2018; 38 Suppl 1:56-63. [PMID: 29427492 DOI: 10.1111/liv.13676] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 12/02/2017] [Indexed: 02/06/2023]
Abstract
Non-alcoholic fatty liver disease is a leading cause of chronic liver disease and can lead to cirrhosis, hepatocellular cancer and end stage liver disease. It is also associated with increased cardiovascular and cancer related morbidity and mortality. The pathogenesis of non-alcoholic fatty liver disease includes metabolic stress to the liver associated with insulin resistance with downstream cell stress from reactive oxygen species and unfolded protein response with activation of inflammatory and fibrotic pathways. There are currently no approved therapies for non-alcoholic fatty liver disease. This review summarizes ongoing efforts to establish the treatment of non-alcoholic steatohepatitis the progressive form of non-alcoholic fatty liver disease. Therapies are currently directed towards improving the metabolic status of the liver, cell stress, apoptosis, inflammation or fibrosis. Several agents are now in pivotal trials and it is expected that the first therapies will be approved in 2-3 years.
Collapse
Affiliation(s)
- Danny Issa
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Vaishali Patel
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Arun J Sanyal
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| |
Collapse
|
794
|
Matey-Hernandez ML, Williams FMK, Potter T, Valdes AM, Spector TD, Menni C. Genetic and microbiome influence on lipid metabolism and dyslipidemia. Physiol Genomics 2018; 50:117-126. [PMID: 29341867 PMCID: PMC5867613 DOI: 10.1152/physiolgenomics.00053.2017] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Disruption in the metabolism of lipids is broadly classified under dyslipidemia and relates to the concentration of lipids in the blood. Dyslipidemia is a predictor of cardio-metabolic disease including obesity. Traditionally, the large interindividual variation has been related to genetic factors and diet. Genome-wide association studies have identified over 150 loci related to abnormal lipid levels, explaining ~40% of the total variation. Part of the unexplained variance has been attributed to environmental factors including diet, but the extent of the dietary contribution remains unquantified. Furthermore, other factors are likely to influence lipid metabolism including the gut microbiome, which plays an important role in the digestion of different dietary components including fats and polysaccharides. Here we describe the contributing role of host genetics and the gut microbiome to dyslipidemia and discuss the potential therapeutic implications of advances in understanding the gut microbiome to the treatment of dyslipidemia.
Collapse
Affiliation(s)
- Maria Luisa Matey-Hernandez
- Department of Twin Research and Genetic Epidemiology, King's College London, St Thomas' Hospital, Westminster Bridge Road, London , United Kingdom
| | - Frances M K Williams
- Department of Twin Research and Genetic Epidemiology, King's College London, St Thomas' Hospital, Westminster Bridge Road, London , United Kingdom
| | - Tilly Potter
- Department of Twin Research and Genetic Epidemiology, King's College London, St Thomas' Hospital, Westminster Bridge Road, London , United Kingdom
| | - Ana M Valdes
- Department of Twin Research and Genetic Epidemiology, King's College London, St Thomas' Hospital, Westminster Bridge Road, London , United Kingdom
- School of Medicine, University of Nottingham, City Hospital Hucknall Rd , Nottingham , United Kingdom
- NIHR Nottingham Biomedical Research Centre , Nottingham , United Kingdom
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, St Thomas' Hospital, Westminster Bridge Road, London , United Kingdom
| | - Cristina Menni
- Department of Twin Research and Genetic Epidemiology, King's College London, St Thomas' Hospital, Westminster Bridge Road, London , United Kingdom
| |
Collapse
|
795
|
Gribble FM, Meek CL, Reimann F. Targeted intestinal delivery of incretin secretagogues-towards new diabetes and obesity therapies. Peptides 2018; 100:68-74. [PMID: 29412834 PMCID: PMC5805852 DOI: 10.1016/j.peptides.2017.11.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 11/16/2017] [Accepted: 11/20/2017] [Indexed: 12/11/2022]
Abstract
A new strategy under development for the treatment of type 2 diabetes and obesity is to mimic some of the effects of bariatric surgery by delivering food-related stimuli to the distal gastrointestinal tract where they should enhance the release of gut hormones such as glucagon-like peptide-1 (GLP-1) and peptideYY (PYY). Methods include inhibition of food digestion and absorption in the upper GI tract, or oral delivery of stimuli in capsules or pelleted form to protect them against gastric degradation. A variety of agents have been tested in humans using capsules, microcapsules or pellets, delivering nutrients, bile acids, fatty acids and bitter compounds. This review examines the outcomes of these different approaches and supporting evidence from intestinal perfusion studies.
Collapse
Affiliation(s)
- Fiona M Gribble
- Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Addenbrooke's Hospital, Box 289, Hills Road, Cambridge, CB2 0QQ, United Kingdom; Department of Clinical Biochemistry, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, United Kingdom.
| | - Claire L Meek
- Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Addenbrooke's Hospital, Box 289, Hills Road, Cambridge, CB2 0QQ, United Kingdom; Department of Clinical Biochemistry, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, United Kingdom
| | - Frank Reimann
- Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Addenbrooke's Hospital, Box 289, Hills Road, Cambridge, CB2 0QQ, United Kingdom; Department of Clinical Biochemistry, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, United Kingdom.
| |
Collapse
|
796
|
Abstract
The obesity epidemic continues to escalate each year in the United States more than anywhere else in the world. The existing pharmaceutical and other nonsurgical treatments for morbid obesity produce suboptimal physiologic outcomes compared with those of Roux-en-Y gastric bypass (RYGB) surgery. RYGB has been the gold standard of bariatric surgery because the beneficial long-term outcomes, which include sustainable weight loss and type 2 diabetes mellitus (T2DM) resolution, are far superior to those obtained with other bariatric surgeries. However, the current understanding of RYGB's mechanisms of actions remains limited and incomplete. There is an urgent need to understand these mechanisms as gaining this knowledge may lead to the development of innovative and less invasive procedures and/or medical devices, which can mirror the favorable outcomes of RYGB surgery. In this review, we highlight current observations of the metabolic and physiologic events following RYGB, with a particular focus on the role of the anatomical reconfiguration of the gastrointestinal tract after RYGB.
Collapse
Affiliation(s)
- Martin L Yarmush
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Burn Hospital for Children, Boston, Massachusetts 02114;
| | - Matthew D'Alessandro
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Burn Hospital for Children, Boston, Massachusetts 02114;
| | - Nima Saeidi
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Burn Hospital for Children, Boston, Massachusetts 02114;
| |
Collapse
|
797
|
Le Bourgot C, Apper E, Blat S, Respondek F. Fructo-oligosaccharides and glucose homeostasis: a systematic review and meta-analysis in animal models. Nutr Metab (Lond) 2018; 15:9. [PMID: 29416552 PMCID: PMC5785862 DOI: 10.1186/s12986-018-0245-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 01/15/2018] [Indexed: 12/25/2022] Open
Abstract
The aim of this systematic review was to assess the effect of fructo-oligosaccharide supplementation on glucose homeostasis. The search process was based on the selection of publications listed in the Pubmed-Medline database until April 2016 to identify studies evaluating the impact of short-chain fructo-oligosaccharides or oligofructose on glucose homeostasis. Twenty-nine trials were included in the systematic review and the meta-analysis was performed on twelve of these papers according to the inclusion criteria. Fasting blood concentrations of glucose and insulin were selected as pertinent criteria of glucose homeostasis for the meta-analysis. The consumption of fructo-oligosaccharides decreased fasting blood glycaemia levels, whatever the metabolic status (healthy, obese or diabetic) and diet (low-fat or high-fat) throughout the experiment. This reduction was linear with prebiotic dose (from 0 to 13% of the feed). Fasting insulinaemia also decreased linearly with fructo-oligosaccharide supplementation but the reduction was only significant in rodents fed a low-fat diet. Potential underlying mechanisms include gut bacterial fermentation of fructo-oligosaccharides to short-chain fatty acids (SCFA) and bacterial modulation of bile acids, both interacting with host metabolism. This systemic review, followed by the meta-analysis, provides evidence that fructo-oligosaccharide supplementation has a significant effect on glucose homeostasis whatever the health status and diet consumed by animals.
Collapse
Affiliation(s)
- Cindy Le Bourgot
- R&D Department, Tereos, ZI et portuaire, 67390 Marckolsheim, France
| | - Emmanuelle Apper
- R&D Department, Tereos, ZI et portuaire, 67390 Marckolsheim, France
| | - Sophie Blat
- 2INRA, INSERM, Univ Rennes 1, Nutrition Metabolisms and Cancer (NuMeCan), Rennes, France
| | | |
Collapse
|
798
|
Yang F, Wang Y, Li G, Xue J, Chen ZL, Jin F, Luo L, Zhou X, Ma Q, Cai X, Li HR, Zhao L. Effects of corilagin on alleviating cholestasis via farnesoid X receptor-associated pathways in vitro and in vivo. Br J Pharmacol 2018; 175:810-829. [PMID: 29235094 DOI: 10.1111/bph.14126] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 11/22/2017] [Accepted: 11/24/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE The aim of this study was to investigate the ameliorative effects of corilagin on intrahepatic cholestasis induced by regulating liver farnesoid X receptor (FXR)-associated pathways in vitro and in vivo. EXPERIMENTAL APPROACH Cellular and animal models were treated with different concentrations of corilagin. In the cellular experiments, FXR expression was up-regulated by either lentiviral transduction or GW4064 treatment and down-regulated by either siRNA technology or treatment with guggulsterones. Real-time PCR and Western blotting were employed to detect the mRNA and protein levels of FXR, SHP1, SHP2, UGT2B4, BSEP, CYP7A1, CYP7B1, NTCP, MRP2 and SULT2A1. Immunohistochemistry was used to examine the expression of BSEP in liver tissues. Rat liver function and pathological changes in hepatic tissue were assessed using biochemical tests and haematoxylin and eosin staining. RESULTS Corilagin increased the mRNA and protein levels of FXR, SHP1, SHP2, UGT2B4, BSEP, MRP2 and SULT2A1, and decreased those of CYP7A1, CYP7B1 and NTCP. After either up- or down-regulating FXR using different methods, corilagin could still increase the mRNA and protein levels of FXR, SHP1, SHP2, UGT2B4, BSEP, MRP2 and SULT2A1 and decrease the protein levels of CYP7A1, CYP7B1 and NTCP, especially when administered at a high concentration. Corilagin also exerted a notable effect on the pathological manifestations of intrahepatic cholestasis, BSEP staining in liver tissues and liver function. CONCLUSIONS AND IMPLICATIONS Corilagin exerts a protective effect in hepatocytes and can prevent the deleterious activities of intrahepatic cholestasis by stimulating FXR-associated pathways.
Collapse
Affiliation(s)
- Fan Yang
- Department of Hepatology, Hubei Provincial Hospital of Chinese Medicine, Wuhan, China
| | - Yao Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Gang Li
- Department of Infectious Diseases, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Juan Xue
- Department of Gastroenterology, Hubei Province Hospital of Integrated Traditional Chinese and Western Medicine, Wuhan, China
| | - Zhi-Lin Chen
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Jin
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University and Shangdong Provincial Key Laboratory of Stem Cells and Neuro-oncology, Jining, Shandong, China
| | - Lei Luo
- School of First Clinical Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Xuan Zhou
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Ma
- School of Life Science, Hubei University, Wuhan, China
| | - Xin Cai
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University and Shangdong Provincial Key Laboratory of Stem Cells and Neuro-oncology, Jining, Shandong, China
| | - Hua-Rong Li
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Zhao
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
799
|
Shapiro H, Kolodziejczyk AA, Halstuch D, Elinav E. Bile acids in glucose metabolism in health and disease. J Exp Med 2018; 215:383-396. [PMID: 29339445 PMCID: PMC5789421 DOI: 10.1084/jem.20171965] [Citation(s) in RCA: 318] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/11/2017] [Accepted: 12/19/2017] [Indexed: 12/11/2022] Open
Abstract
Bile acids (BAs) are cholesterol-derived metabolites that facilitate the intestinal absorption and transport of dietary lipids. Recently, BAs also emerged as pivotal signaling molecules controlling glucose, lipid, and energy metabolism by binding to the nuclear hormone farnesoid X receptor (FXR) and Takeda G protein receptor 5 (TGR5) in multiple organs, leading to regulation of intestinal incretin secretion, hepatic gluconeogenesis, glycogen synthesis, energy expenditure, inflammation, and gut microbiome configuration. Alterations in BA metabolism and signaling are associated with obesity and type 2 diabetes mellitus (T2DM), whereas treatment of T2DM patients with BA sequestrants, or bariatric surgery in morbidly obese patients, results in a significant improvement in glycemic response that is associated with changes in the BA profile and signaling. Herein, we review the roles of BAs in glucose metabolism in health and disease; highlight the limitations, unknowns, and challenges in understanding the impact of BAs on the glycemic response; and discuss how this knowledge may be harnessed to develop innovative therapeutic approaches for the treatment of hyperglycemia and diabetes.
Collapse
Affiliation(s)
- Hagit Shapiro
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Daniel Halstuch
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Elinav
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
800
|
TGR5 signalling promotes mitochondrial fission and beige remodelling of white adipose tissue. Nat Commun 2018; 9:245. [PMID: 29339725 PMCID: PMC5770450 DOI: 10.1038/s41467-017-02068-0] [Citation(s) in RCA: 181] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 11/03/2017] [Indexed: 01/03/2023] Open
Abstract
Remodelling of energy storing white fat into energy expending beige fat could be a promising strategy to reduce adiposity. Here, we show that the bile acid-responsive membrane receptor TGR5 mediates beiging of the subcutaneous white adipose tissue (scWAT) under multiple environmental cues including cold exposure and prolonged high-fat diet feeding. Moreover, administration of TGR5-selective bile acid mimetics to thermoneutral housed mice leads to the appearance of beige adipocyte markers and increases mitochondrial content in the scWAT of Tgr5+/+ mice but not in their Tgr5−/− littermates. This phenotype is recapitulated in vitro in differentiated adipocytes, in which TGR5 activation increases free fatty acid availability through lipolysis, hence fuelling β-oxidation and thermogenic activity. TGR5 signalling also induces mitochondrial fission through the ERK/DRP1 pathway, further improving mitochondrial respiration. Taken together, these data identify TGR5 as a druggable target to promote beiging with potential applications in the management of metabolic disorders. White adipose tissue can undergo a process of beiging and acquire functional characteristics similar to brown adipose tissue, including the ability to dissipate energy via uncoupled respiration. Here, Velazquez-Villegas et al. show that activation of the bile acid membrane receptor, TGR5, leads to white adipocyte beiging by promoting mitochondrial fission.
Collapse
|