Please wait a minute...
Frontiers of Medicine

ISSN 2095-0217

ISSN 2095-0225(Online)

CN 11-5983/R

Postal Subscription Code 80-967

2018 Impact Factor: 1.847

Front. Med.    2018, Vol. 12 Issue (6) : 608-623    https://doi.org/10.1007/s11684-018-0644-x
REVIEW
Bile acids and their effects on diabetes
Cynthia Rajani1, Wei Jia1,2()
1. University of Hawaii Cancer Center, Honolulu, HI 96813, USA
2. Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
 Download: PDF(661 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Diabetes is a widespread, rapidly increasing metabolic disease that is driven by hyperglycemia. Early glycemic control is of primary importance to avoid vascular complications including development of retinal disorders leading to blindness, end-stage renal disease, and accelerated atherosclerosis with a higher risk of myocardial infarction, stroke and limb amputations. Even after hyperglycemia has been brought under control, “metabolic memory,” a cluster of irreversible metabolic changes that allow diabetes to progress, may persist depending on the duration of hyperglycemia. Manipulation of bile acid (BA) receptors and the BA pool have been shown to be useful in establishing glycemic control in diabetes due to their ability to regulate energy metabolism by binding and activating nuclear transcription factors such as farnesoid X receptor (FXR) in liver and intestine as well as the G-protein coupled receptor, TGR5, in enteroendocrine cells and pancreatic β-cells. The downstream targets of BA activated FXR, FGF15/21, are also important for glucose/insulin homeostasis. In this review we will discuss the effect of BAs on glucose and lipid metabolism and explore recent research on establishing glycemic control in diabetes through the manipulation of BAs and their receptors in the liver, intestine and pancreas, alteration of the enterohepatic circulation, bariatric surgery and alignment of circadian rhythms.

Keywords bile acids      metabolic memory      diabetes      circadian rhythm      bariatric surgery     
Corresponding Author(s): Wei Jia   
Just Accepted Date: 06 August 2018   Online First Date: 16 October 2018    Issue Date: 03 December 2018
 Cite this article:   
Cynthia Rajani,Wei Jia. Bile acids and their effects on diabetes[J]. Front. Med., 2018, 12(6): 608-623.
 URL:  
https://academic.hep.com.cn/fmd/EN/10.1007/s11684-018-0644-x
https://academic.hep.com.cn/fmd/EN/Y2018/V12/I6/608
Fig.1  The Effects of hyperglycemia on the development of metabolic memory. Excess glucose can exceed glycogen storage capacity and use for energy. When this happens, several alternative glucose utilizing pathways become active. The polyol or aldose reductase pathway converts glucose to sorbitol with concurrent production of additional NADH which when reacted with NADH oxidase leads to the production of excess ROS ultimately causing mitochondrial dysfunction. Sorbitol can also be converted to fructose in the polyol pathway and enter the glycolytic pathway to become F-6-P and then onto F-1,6-bis-P which in turn can become converted to GA3P and then methylglyoxal, a precursor for AGEs. Alternatively, F-1,6-bis-P can be biotransformed into DHAP which then enters 2 cycles, one which leads back to F-1,6-bis-P and more AGEs and another which leads to synthesis of G-3-P which can go on to be converted to DAG. DAG blocks AKT→ IRS1/2 leading to decreased insulin sensitivity. In the glycolytic pathway, G-6-P can be diverted to the hexosamine pathway which leads to an increase in O-GlcNAc proteins that in turn can cause increased insulin resistance, gluconeogenesis and lipogenesis [2,6,810]. Abbreviations: hexokinase (HK), glucose-6-phosphate (G-6-P), fructose-6-phosphate (F-6-P), fructose-1,6-bis-phosphate (F-1,6-bis-P), glyceraldehydes-3-phosphate (GA3P), advanced glycosylation end products (AGEs), reactive oxygen species (ROS), dihydroxyacetone phosphate (DHAP), O-linked-N-acetylglucosamine (O-GlcNAc), glucosamine-fructose-6-phosphate aminotransferase (GFAT), glucose-3-phosphate (G3P), glucose-3-phosphate dehydrogenase (G-3-P DH), diacylglycerol (DAG), protein kinase C (PKC), protein kinase B (AKT), insulin receptor substrate 1/2 (IRS1/2), insulin receptor (IR), forkhead box protein O1 (FoxO1), carbohydrate response element binding protein (ChREBP), phosphoenolate pyruvate carboxylase (PEPCK).
Fig.2  The effects of BAs on glucose and lipid metabolism in the normal liver. BAs are considered positive regulators of metabolism in the liver. BA activation of FXR causes its translocation to the nucleus where it forms a heterodimer with RXR. Several signaling pathways are initiated as a result. The co-factor, SHP becomes active in binding the transcription factor, HNF4 which in turn, releases its co-factor CBP. This results in inhibition of the transcription of two important enzymes in gluconeogenesis, PEPCK and FBP1. SHP also can bind to the transcription factor FoxO1 which in turn releases its co-factor, CBP with the result that the enzyme G6Pase does not get expressed. In this way BA activated FXR inhibits gluconeogenesis and prevents release of more glucose into the circulation. BA activated FXR also modulates the transcription factor LXR. LXR is a positive regulator of the transcription factors, ChREBP and SREBP1c which act synergistically to control DNL and glycolysis. Both insulin and glucose can also activate LXR directly. FXR is a negative regulator of LXR and thus BAs via FXR have the effect of decreasing TG formation and accumulation in the liver. Decreased glycolysis leading to pyruvate may also lead to increased shunting into the glycogenesis pathway. BA activated FXR also increases expression of the transcription factor PPARα which leads to increased transcription of genes important for β-oxidation of FAs, therefore increasing the use of FAs for energy rather for storage. PPARα activation also has the effect of stimulating the expression of FGF21 which has important ant-diabetic effects in adipose (Fig. 3) tissue. Lastly, FXR controls the transcription of genes important in lipid handling and transport such as increased expression of ApoCII which increases the activity of LPL and therefore more FAs are taken up by adipose, thus lowering plasma TGs. FXR also causes increased production of ApoA5 which acts to reduce hepatic uptake of TGs. FXR last of all, causes decreased expression of ApoCIII which has the effect of increasing FA uptake by the liver and thus lowers serum FA levels [11,2427]. Abbreviations: bile acid (BA), farnesoid X receptor (FXR), glucose receptor 2 (GLUT2), glucokinase (GK), glucose-6-phosphate (G6P), phosphoenolpyruvate (PEP), pyruvate kinase (PK), acetyl-CoA carboxylase (ACC), fatty acid synthase (FAS), steroyl-CoA-desaturase-1 (SCD1), liver X receptor (LXR), sterol response element binding protein-1c (SREBP1c), carbohydrate response element binding protein (ChREBP), retinoic acid receptor (RXR), fibroblast growth factor (FGF21), small heterodimer partner (SHP), hepatic nuclear factor-4 (HNF4), cAMP response element binding protein (CBP), forkhead transcription factor FoxO1 (FoxO1), glucose-6-phosphatase (G6Pase), phosphoenolatepyruvate carboxylase (PEPCK), fructose-1,6-bis-phosphatase (FBP1).
Fig.3  The effect of BAs on FGF19/21 and ceramide signaling relative to lipid and glucose metabolism. BA activated FXR has tissue specific effects with respect to hormone production. In the intestine, BAs activate FXR to cause production of FGF21 which is then secreted from the liver and binds to its receptor, FGFR4/β-Klotho on WAT causing activation of the ERK1/2→ RSK →Elk1/SRF pathway which in turn, causes increased expression of GLUT1 receptors on the adipocyte surface thus reducing hyperglycemia by increasing glucose uptake into the cell. FGF21 in WAT also causes increased secretion of adiponectin which has the effect of decreasing serum ceramide levels and increasing the number of beige of adipocytes thereby, increasing the energy utilization capabilities of the adipose tissue to help fight obesity. In the liver, FGF21 stimulates β-oxidation of FAs, as well as, increased ketogenesis. BA activation of FXR in the intestine targets two genes for enzymes important for the synthesis of ceramide, Smpd3 and Sptlc2, leading to increased serum levels of ceramide. In addition to the previously discussed effect on WAT, ceramide, when it is taken up by the liver becomes an activator of several signaling pathways which together act to block insulin signaling, hence, insulin resistance results. Ceramide also directly acts on the transcription factor, SREBP-1c, to increase DNL and lipid synthesis. The other consequence of intestinal FXR activation is the production on FGF19 which is secreted and then binds to its receptor, FGFR4/β-Klotho, in the liver where it acts to decrease BA synthesis and activates a hepatic ERK1/2 signaling pathway which results in increased protein synthesis, including GLUT1 receptors to increase glucose uptake and also increased glycogen synthesis to increase storage of glucose as glycogen. Both effects act to decrease hyperglycemia [28,32,3638]. Abbreviations: bile acid (BA), cholic acid (CA), chenodeoxycholic acid (CDCA), farnesoid X receptor (FXR), retinoid X receptor (RXR), fibroblast growth factor 19/21 (FGF19/21), insulin receptor (IR), insulin receptor substrate 1/2 (IRS1/2), inhibitor of nuclear factor kB kinase subunit β (IKK2), c-Jun N-terminal kinase (JNK), protein kinase C ζ (PKCζ), fatty acid (FA), protein kinase B (AKT), sterol response element binding protein-1c (SREBP1c), carbohydrate response element binding protein (ChREBP), S6 ribosomal protein (S6), eukaryotic translation initiator factor-4B (elF-4B), glycogen synthase kinase 3 (GSK3), glycogen synthase (GS), ribosomal S6 kinase (RSK), extracellular signal-related kinase 1/2 (ERK1/2), fibroblast growth factor receptor-4 (FGFR4), ETS domain containing protein-1 (Elk1), serum response factor (SRF), bile salt hydrolase (BSH), white adipose tissue (WAT), GLUT1 glucose transporter (GLUT1).
FGF-19 1° Targets 2° Targets Cellular effects Metabolic effects
Originates from BA activation of intestinal FXR Hepatic FGFR4/β-Klotho receptor complex ERK1/2 ↓Hepatic BA synthesis ↓BA concentrations in liver and gut
RSK ↑Hepatic GLUT1 receptors ↑Hepatic glucose uptake
↓Blood sugar
GSK3 ↑Glycogen synthase ↑Glycogen synthesis/storage from glucose
↓Blood sugar
FGF-21 1° Targets 2° Targets Cellular effects Metabolic effects
Originates from BA activation of hepatic FXR Adipose FGFR4/β-Klotho receptor complex ERK1/2+ RSK ↑Adipose GLUT1
receptors
↑Adipose glucose uptake
↓Blood sugar
↑Adiponectin secretion ↓Serum ceramide concentration
↑Number of beige adipocytes ↑Energy utilization by adipocytes
↓Lipid storage/obesity
PPARa/FGF21 synergism with PPARα agonists in WAT Upregulation of Ucp1 and Pgc1α in WAT but not BAT ↑Production of beige adipocytes in WAT ↑β-oxidation of FAs
↑Ketogenesis
↓Lipid storage/obesity
↓Blood sugar
↓Blood insulin
Tab.1  Effects of FGF-19/21 on metabolism [32, 36, 37, 42]
Tissue FGF15 Conclusion for FGF15 FGF21 Conclusion for FGF21
Liver KO (KlbAlb mice)/ treatments ↓CYP7A1
↓FGF15/βKlotho-FGFR4 signaling
FGF15/FGFR4 essential for BA homeostasis No acute effect on CYP7A1, ERK1/2 No acute, direct effect on hepatocytes
HFD (7 days) DIO both for KO and control with no changes in glucose production, uptake despite elevated FGF15 Hepatic FGF15 not essential for systemic glucose metabolism
2-week administration of FGF15/21 ↓Body weight, serum glucose, hepatic TG Improved metabolic parameters do not require FGF15/21 in liver ↓Body weight, serum glucose, hepatic TG Improved metabolic parameters do not require FGF15/21 in liver
Adipose KO (BAT,WAT)
Klbadipo mice/ longterm treatment FGF15/21
↑Whole body insulin sensitivity
↓Body weight, plasma insulin/glucose, hepatic TGs
↓Hepatic DAG for both control and model mice
↑Expression of Dusp4 in control mice only
The effects of these hormones do not require direct action on the adipocytes in Klbadipo mice but do directly affect ERK1/2 signaling in control mice ↑Whole body insulin sensitivity
↓Body weight, plasma insulin/glucose, hepatic TGs
↓Hepatic DAG for both control and model mice
↑Expression of Dusp4 in control mice only
The effects of these hormones do not require direct action on the adipocytes in Klbadipo mice but do directly affect ERK1/2 signaling in control mice
NS KO (KlbCamk2a mice)
Longterm treatment FGF15
Only control mice showed weight loss and improved glycemic control
Sympathetic nerve activity in BAT was ↑ in a dose dependent way with FGF15 treatment
FGF15 acts on the NS in a way similar to FGF21 (previous study)[45]
Administration of bFKB1 Ab to the FGFR1 receptor KlbCamk2a mice were resistant to weight loss and improved gylycemic control This confirmed the result in the FGF15/21 model βKlotho/FGFR1 complexes are essential in the NS
Tab.2  Effects of tissue-specific β-Klotho loss of function in mouse liver, adipose and nervous system on the nervous system relative to a floxed control [19]
Fig.4  The effects of BA activated TGR5 signaling in enteroendocrine cells (EECs) and in pancreatic β-cells. (A) EECc produce and secrete important hormones that affect energy metabolism and preserve pancreatic β-cell function. In EECs, TGR5 and GPR119 are coupled to Gαs G-proteins while GPR40 is coupled to a Gαq G-protein. Secondary BAs are agonists for TGR5 while 2-MAG and LCFAs are agonists for GPR119 and GPR40, respectively. All three GPCRs promote secretion of GLP-1, GIP and PYY, incretins that have important effects on glucose homeostasis. Gαs protein coupling to TGR5 and GPR119 results in the recruitment of adenyl cyclase which subsequently activates cAMP to increase intracellular Ca2+ via PKA or Epac pathways and ultimately increase secretion of GLP-1 and other incretins. Gαq protein coupling to GPR40 activates an alternate pathway to increase intracellular Ca2+ , PLC → IP3. Furthermore, Gαq signaling via PLC can activate DAG →PKC to cause increased incretin secretion via a Ca2+ independent pathway. It was determined that both Gαs and Gαq coupling to GPCRS worked in a synergistic way to increase intracellular Ca2+ levels which in turn stimulated vesicle fusion and increased secretion of incretins. (B) In the pancreatic β-cell, the BA TUDCA was used to activate TGR5 and this was found not only to increase the secretion of insulin via Ca2+ stimulated exocytosis using the same mechanism as seen in EECs for increased incretin secretion but TGR5 activation also led to activation of the CREB→ Bcl2/BclXL pathway to decrease apoptosis and additionally, activation of the CREB → IRS-2 → PDX pathway which leads to increased maturation of β-cells. The net result of these pathway activations is increased β-cell mass. TUDCA activation of TGR5 also activates ATF-4 → Gadd34 →elF2α signaling to cause increased protein translation which acts to decrease ER stress [46−52]. Abbreviations: lithocholic acid (LCA), deoxycholic acid (DCA), adenosine triphosphate (ATP), cyclic adenosine monophosphate (cAMP), protein kinase A (PKA), guanine nucleotide exchange factor (Epac), ryanodine receptor (RYR), long chain fatty acid (LCFA), inositol triphosphate (IP3), inositol triphosphate receptor (IP3R), endoplasmic reticulum (ER), diacylglycerol (DAG), phospholipase C (PLC), protein kinase C (PKC), peptide YY (PYY), glucose dependent insulintropic polypeptide protein (GIP), glucagon-like peptide-1 (GLP-1), hydroxysterol dehydrogenase (HSDH), chenodeoxycholic acid (CDCA), tauroursodeoxycholic acid (TUDCA), cAMP response element binding protein (CREB), activating transcription factor-4 (ATF-4), B cell lymphoma-2 (Bcl-2), B cell lymphoma extra large (BclXL), pancreatic and duodenal homeobox-1 (PDX1), insulin receptor substrate-2 (IRS2), growth arrest and DNA damage induced protein-34 (Gadd34), eukaryotic initiation factor 2α (elF2α).
Fig.5  The circadian control of BA synthesis, lipogenesis and the Octn1 transporter. In hepatocytes, there exists a circadian core clock that consists of an autoregulated feedback loop of rhythmically expressed genes that oscillate within a 24 h period. Two important genes, Clock and Bmal1 comprise the forward segment of the clock loop. CLOCK and BMAL1 proteins form a heterodimer which then bind to the DNA response element, E-box, to cause the transcription of Per and Cry genes. PER and CRY proteins then re-enter the nucleus and inhibit CLOCK:BMAL1 protein activity to reduce their own transcription. There is further regulation of the feedback loop by the nuclear hormone receptors, Rev-erbα which negatively regulates and RORα that positively regulates Bmal1 transcription [70]. REV-ERBα effects on BA metabolism include feeding independent circadian control of SREBP2, SREBP1c, the transcription factors for enzymes involved in lipogenesis and cholesterol-7α-hydroxylase (CYP7A1), the rate-limiting enzyme for BA synthesis in the liver. Transcription of a key protein, Insig2 is blocked by REV-ERBα and this causes increased release of SREBP from the ER and allows it to translocate into the nucleus. The circadian rhythm of SREBP2 causes corresponding changes in oxysterol production which are known to regulate LXR and increase transcription of Cyp7a1 [68]. Both PPARα and the co-activator, peroxisome proliferator-activated receptor gamma co-activator 1-α (PGC1α) modulate Bmal1 transcription [64,70]. PPARα is a positive regulator of the Octn1 transporter via control of transcription of the Slc22a4 gene [71]. In addition, certain nutrient-sensitive signaling pathways such as SIRT1 and AMPK couple metabolic flux to the circadian cycle [70]. Abbreviations: circadian locomotor output cycles kaput (Clock), brain and muscle Arnt-like 1 (Bmal1), period (Per), cryptochrome (Cry), reverse-erythroblastosis α (Rev-erbα), retinoic acid-related orphan receptor α (RORα), cholesterol-7α-hydroxylase (CYP7A1), sterol response element binding protein (SREBP). liver X receptor (LXR, peroxisome proliferator-activated receptor α(PPARα), peroxisome proliferator-activated receptor gamma co-activator 1-α (PGC1α), insulin-induced gene-2 (Insig2), SREBP-cleavage activating protein (SCAP), adenosine monophosphate-activated protein kinase (AMPK), nicotinamide adenine dinucleotide (NAD), sirtuin 1 (SIRT1), casein kinase 1(CK1), F-box/LRR repeat protein 3 (FBLX3), organic cation transporter novel type-1 (Octn1).
1 Abramowicz M, Zuccotti G. Drugs for diabetes. Treat Guidel Med Lett 2005; 3(36): 57–62
pmid: 16012392
2 Ceriello A. The emerging challenge in diabetes: the “metabolic memory”. Vascul Pharmacol 2012; 57(5-6): 133–138
https://doi.org/10.1016/j.vph.2012.05.005 pmid: 22609133
3 Ceriello A, Esposito K, Piconi L, Ihnat MA, Thorpe JE, Testa R, Boemi M, Giugliano D. Oscillating glucose is more deleterious to endothelial function and oxidative stress than mean glucose in normal and type 2 diabetic patients. Diabetes 2008; 57(5): 1349–1354
https://doi.org/10.2337/db08-0063 pmid: 18299315
4 Ceriello A, Esposito K, Ihnat M, Thorpe J, Giugliano D. Long-term glycemic control influences the long-lasting effect of hyperglycemia on endothelial function in type 1 diabetes. J Clin Endocrinol Metab 2009; 94(8): 2751–2756
https://doi.org/10.1210/jc.2009-0762 pmid: 19491221
5 Nathan DM, Cleary PA, Backlund JY, Genuth SM, Lachin JM, Orchard TJ, Raskin P, Zinman B; Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications (DCCT/EDIC) Study Research Group. Intensive diabetes treatment and cardiovascular disease in patients with type 1 diabetes. N Engl J Med 2005; 353(25): 2643–2653
https://doi.org/10.1056/NEJMoa052187 pmid: 16371630
6 Brownlee M. Biochemistry and molecular cell biology of diabetic complications. Nature 2001; 414(6865): 813–820
https://doi.org/10.1038/414813a pmid: 11742414
7 Foufelle F, Ferré P. New perspectives in the regulation of hepatic glycolytic and lipogenic genes by insulin and glucose: a role for the transcription factor sterol regulatory element binding protein-1c. Biochem J 2002; 366(Pt 2): 377–391
https://doi.org/10.1042/bj20020430 pmid: 12061893
8 Banerjee PS, Lagerlöf O, Hart GW. Roles of O-GlcNAc in chronic diseases of aging. Mol Aspects Med 2016; 51: 1–15
https://doi.org/10.1016/j.mam.2016.05.005 pmid: 27259471
9 Hanssen NM, Beulens JW, van Dieren S, Scheijen JL, van der A DL, Spijkerman AM, van der Schouw YT, Stehouwer CD, Schalkwijk CG. Plasma advanced glycation end products are associated with incident cardiovascular events in individuals with type 2 diabetes: a case-cohort study with a median follow-up of 10 years (EPIC-NL). Diabetes 2015; 64(1): 257–265
https://doi.org/10.2337/db13-1864 pmid: 24848072
10 Guinez C, Filhoulaud G, Rayah-Benhamed F, Marmier S, Dubuquoy C, Dentin R, Moldes M, Burnol AF, Yang X, Lefebvre T, Girard J, Postic C. O-GlcNAcylation increases ChREBP protein content and transcriptional activity in the liver. Diabetes 2011; 60(5): 1399–1413
https://doi.org/10.2337/db10-0452 pmid: 21471514
11 Li T, Chiang JY. Bile acid signaling in metabolic disease and drug therapy. Pharmacol Rev 2014; 66(4): 948–983
https://doi.org/10.1124/pr.113.008201 pmid: 25073467
12 Kong B, Wang L, Chiang JY, Zhang Y, Klaassen CD, Guo GL. Mechanism of tissue-specific farnesoid X receptor in suppressing the expression of genes in bile-acid synthesis in mice. Hepatology 2012; 56(3): 1034–1043
https://doi.org/10.1002/hep.25740 pmid: 22467244
13 Thomas C, Pellicciari R, Pruzanski M, Auwerx J, Schoonjans K. Targeting bile-acid signalling for metabolic diseases. Nat Rev Drug Discov 2008; 7(8): 678–693
https://doi.org/10.1038/nrd2619 pmid: 18670431
14 Trauner M, Boyer JL. Bile salt transporters: molecular characterization, function, and regulation. Physiol Rev 2003; 83(2): 633–671
https://doi.org/10.1152/physrev.00027.2002 pmid: 12663868
15 Zhang YK, Guo GL, Klaassen CD. Diurnal variations of mouse plasma and hepatic bile acid concentrations as well as expression of biosynthetic enzymes and transporters. PLoS One 2011; 6(2): e16683
https://doi.org/10.1371/journal.pone.0016683 pmid: 21346810
16 Ridlon JM, Kang DJ, Hylemon PB. Bile salt biotransformations by human intestinal bacteria. J Lipid Res 2006; 47(2): 241–259
https://doi.org/10.1194/jlr.R500013-JLR200 pmid: 16299351
17 Dawson PA, Lan T, Rao A. Bile acid transporters. J Lipid Res 2009; 50(12): 2340–2357
https://doi.org/10.1194/jlr.R900012-JLR200 pmid: 19498215
18 Meier PJ, Stieger B. Bile salt transporters. Annu Rev Physiol 2002; 64(1): 635–661
https://doi.org/10.1146/annurev.physiol.64.082201.100300 pmid: 11826283
19 Lan T, Morgan DA, Rahmouni K, Sonoda J, Fu X, Burgess SC, Holland WL, Kliewer SA, Mangelsdorf DJ. FGF19, FGF21, and an FGFR1/β-Klotho-activating antibody act on the nervous system to regulate body weight and glycemia. Cell Metab 2017; 26(5): 709–718.e3
https://doi.org/10.1016/j.cmet.2017.09.005 pmid: 28988823
20 Rizza RA. Pathogenesis of fasting and postprandial hyperglycemia in type 2 diabetes: implications for therapy. Diabetes 2010; 59(11): 2697–2707
https://doi.org/10.2337/db10-1032 pmid: 20705776
21 Chung ST, Hsia DS, Chacko SK, Rodriguez LM, Haymond MW. Increased gluconeogenesis in youth with newly diagnosed type 2 diabetes. Diabetologia 2015; 58(3): 596–603
https://doi.org/10.1007/s00125-014-3455-x pmid: 25447079
22 Yamagata K, Daitoku H, Shimamoto Y, Matsuzaki H, Hirota K, Ishida J, Fukamizu A. Bile acids regulate gluconeogenic gene expression via small heterodimer partner-mediated repression of hepatocyte nuclear factor 4 and Foxo1. J Biol Chem 2004; 279(22): 23158–23165
https://doi.org/10.1074/jbc.M314322200 pmid: 15047713
23 Bechmann LP, Hannivoort RA, Gerken G, Hotamisligil GS, Trauner M, Canbay A. The interaction of hepatic lipid and glucose metabolism in liver diseases. J Hepatol 2012; 56(4): 952–964
https://doi.org/10.1016/j.jhep.2011.08.025 pmid: 22173168
24 Dentin R, Girard J, Postic C. Carbohydrate responsive element binding protein (ChREBP) and sterol regulatory element binding protein-1c (SREBP-1c): two key regulators of glucose metabolism and lipid synthesis in liver. Biochimie 2005; 87(1): 81–86
https://doi.org/10.1016/j.biochi.2004.11.008 pmid: 15733741
25 Ip E, Farrell GC, Robertson G, Hall P, Kirsch R, Leclercq I. Central role of PPARα-dependent hepatic lipid turnover in dietary steatohepatitis in mice. Hepatology 2003; 38(1): 123–132
https://doi.org/10.1053/jhep.2003.50307 pmid: 12829994
26 Denechaud PD, Dentin R, Girard J, Postic C. Role of ChREBP in hepatic steatosis and insulin resistance. FEBS Lett 2008; 582(1): 68–73
https://doi.org/10.1016/j.febslet.2007.07.084 pmid: 17716660
27 Zhang Y, Lee FY, Barrera G, Lee H, Vales C, Gonzalez FJ, Willson TM, Edwards PA. Activation of the nuclear receptor FXR improves hyperglycemia and hyperlipidemia in diabetic mice. Proc Natl Acad Sci USA 2006; 103(4): 1006–1011
https://doi.org/10.1073/pnas.0506982103 pmid: 16410358
28 Gonzalez FJ, Jiang C, Patterson AD. An Intestinal microbiota-farnesoid X receptor axis modulates metabolic disease. Gastroenterology 2016; 151(5): 845–859
https://doi.org/10.1053/j.gastro.2016.08.057 pmid: 27639801
29 Turpin SM, Nicholls HT, Willmes DM, Mourier A, Brodesser S, Wunderlich CM, Mauer J, Xu E, Hammerschmidt P, Brönneke HS, Trifunovic A, LoSasso G, Wunderlich FT, Kornfeld JW, Blüher M, Krönke M, Brüning JC. Obesity-induced CerS6-dependent C16:0 ceramide production promotes weight gain and glucose intolerance. Cell Metab 2014; 20(4): 678–686
https://doi.org/10.1016/j.cmet.2014.08.002 pmid: 25295788
30 Jiang C, Xie C, Lv Y, Li J, Krausz KW, Shi J, Brocker CN, Desai D, Amin SG, Bisson WH, Liu Y, Gavrilova O, Patterson AD, Gonzalez FJ. Intestine-selective farnesoid X receptor inhibition improves obesity-related metabolic dysfunction. Nat Commun 2015; 6(1): 10166
https://doi.org/10.1038/ncomms10166 pmid: 26670557
31 Li F, Jiang C, Krausz KW, Li Y, Albert I, Hao H, Fabre KM, Mitchell JB, Patterson AD, Gonzalez FJ. Microbiome remodelling leads to inhibition of intestinal farnesoid X receptor signalling and decreased obesity. Nat Commun 2013; 4(1): 2384
https://doi.org/10.1038/ncomms3384 pmid: 24064762
32 Kir S, Beddow SA, Samuel VT, Miller P, Previs SF, Suino-Powell K, Xu HE, Shulman GI, Kliewer SA, Mangelsdorf DJ. FGF19 as a postprandial, insulin-independent activator of hepatic protein and glycogen synthesis. Science 2011; 331(6024): 1621–1624
https://doi.org/10.1126/science.1198363 pmid: 21436455
33 Stefano GB, Challenger S, Kream RM. Hyperglycemia-associated alterations in cellular signaling and dysregulated mitochondrial bioenergetics in human metabolic disorders. Eur J Nutr 2016; 55(8): 2339–2345
https://doi.org/10.1007/s00394-016-1212-2 pmid: 27084094
34 Whelan SA, Dias WB, Thiruneelakantapillai L, Lane MD, Hart GW. Regulation of insulin receptor substrate 1 (IRS-1)/AKT kinase-mediated insulin signaling by O-linked β-N-acetylglucosamine in 3T3-L1 adipocytes. J Biol Chem 2010; 285(8): 5204–5211
https://doi.org/10.1074/jbc.M109.077818 pmid: 20018868
35 Housley MP, Rodgers JT, Udeshi ND, Kelly TJ, Shabanowitz J, Hunt DF, Puigserver P, Hart GW. O-GlcNAc regulates FoxO activation in response to glucose. J Biol Chem 2008; 283(24): 16283–16292
https://doi.org/10.1074/jbc.M802240200 pmid: 18420577
36 Holland WL, Adams AC, Brozinick JT, Bui HH, Miyauchi Y, Kusminski CM, Bauer SM, Wade M, Singhal E, Cheng CC, Volk K, Kuo MS, Gordillo R, Kharitonenkov A, Scherer PE. An FGF21-adiponectin-ceramide axis controls energy expenditure and insulin action in mice. Cell Metab 2013; 17(5): 790–797
https://doi.org/10.1016/j.cmet.2013.03.019 pmid: 23663742
37 Ge X, Chen C, Hui X, Wang Y, Lam KS, Xu A. Fibroblast growth factor 21 induces glucose transporter-1 expression through activation of the serum response factor/Ets-like protein-1 in adipocytes. J Biol Chem 2011; 286(40): 34533–34541
https://doi.org/10.1074/jbc.M111.248591 pmid: 21846717
38 Copple BL, Li T. Pharmacology of bile acid receptors: evolution of bile acids from simple detergents to complex signaling molecules. Pharmacol Res 2016; 104: 9–21
https://doi.org/10.1016/j.phrs.2015.12.007 pmid: 26706784
39 Monnier L, Lapinski H, Colette C. Contributions of fasting and postprandial plasma glucose increments to the overall diurnal hyperglycemia of type 2 diabetic patients: variations with increasing levels of HbA(1c). Diabetes Care 2003; 26(3): 881–885
https://doi.org/10.2337/diacare.26.3.881 pmid: 12610053
40 Sonne DP, van Nierop FS, Kulik W, Soeters MR, Vilsbøll T, Knop FK. Postprandial plasma concentrations of individual bile acids and FGF-19 in patients with type 2 diabetes. J Clin Endocrinol Metab 2016; 101(8): 3002–3009
https://doi.org/10.1210/jc.2016-1607 pmid: 27270475
41 Mueller M, Thorell A, Claudel T, Jha P, Koefeler H, Lackner C, Hoesel B, Fauler G, Stojakovic T, Einarsson C, Marschall HU, Trauner M. Ursodeoxycholic acid exerts farnesoid X receptor-antagonistic effects on bile acid and lipid metabolism in morbid obesity. J Hepatol 2015; 62(6): 1398–1404
https://doi.org/10.1016/j.jhep.2014.12.034 pmid: 25617503
42 Goto T, Hirata M, Aoki Y, Iwase M, Takahashi H, Kim M, Li Y, Jheng HF, Nomura W, Takahashi N, Kim CS, Yu R, Seno S, Matsuda H, Aizawa-Abe M, Ebihara K, Itoh N, Kawada T. The hepatokine FGF21 is crucial for peroxisome proliferator-activated receptor-α agonist-induced amelioration of metabolic disorders in obese mice. J Biol Chem 2017; 292(22): 9175–9190
https://doi.org/10.1074/jbc.M116.767590 pmid: 28404815
43 Reitman ML, Gavrilova O. A-ZIP/F-1 mice lacking white fat: a model for understanding lipoatrophic diabetes. Int J Obes Relat Metab Disord 2000; 24 (Suppl 4): S11–14
pmid: 11126232
44 Katafuchi T, Esterházy D, Lemoff A, Ding X, Sondhi V, Kliewer SA, Mirzaei H, Mangelsdorf DJ. Detection of FGF15 in plasma by stable isotope standards and capture by anti-peptide antibodies and targeted mass spectrometry. Cell Metab 2015; 21(6): 898–904
https://doi.org/10.1016/j.cmet.2015.05.004 pmid: 26039452
45 Owen BM, Ding X, Morgan DA, Coate KC, Bookout AL, Rahmouni K, Kliewer SA, Mangelsdorf DJ. FGF21 acts centrally to induce sympathetic nerve activity, energy expenditure, and weight loss. Cell Metab 2014; 20(4): 670–677
https://doi.org/10.1016/j.cmet.2014.07.012 pmid: 25130400
46 Psichas A, Reimann F, Gribble FM. Gut chemosensing mechanisms. J Clin Invest 2015; 125(3): 908–917
https://doi.org/10.1172/JCI76309 pmid: 25664852
47 van Nierop FS, Scheltema MJ, Eggink HM, Pols TW, Sonne DP, Knop FK, Soeters MR. Clinical relevance of the bile acid receptor TGR5 in metabolism. Lancet Diabetes Endocrinol 2017; 5(3): 224–233
https://doi.org/10.1016/S2213-8587(16)30155-3 pmid: 27639537
48 Hauge M, Ekberg JP, Engelstoft MS, Timshel P, Madsen AN, Schwartz TW. Gq and Gs signaling acting in synergy to control GLP-1 secretion. Mol Cell Endocrinol 2017; 449: 64–73
pmid: 27908836
49 Vettorazzi JF, Ribeiro RA, Borck PC, Branco RC, Soriano S, Merino B, Boschero AC, Nadal A, Quesada I, Carneiro EM. The bile acid TUDCA increases glucose-induced insulin secretion via the cAMP/PKA pathway in pancreatic β cells. Metabolism 2016; 65(3): 54–63
https://doi.org/10.1016/j.metabol.2015.10.021 pmid: 26892516
50 Thomas C, Gioiello A, Noriega L, Strehle A, Oury J, Rizzo G, Macchiarulo A, Yamamoto H, Mataki C, Pruzanski M, Pellicciari R, Auwerx J, Schoonjans K. TGR5-mediated bile acid sensing controls glucose homeostasis. Cell Metab 2009; 10(3): 167–177
https://doi.org/10.1016/j.cmet.2009.08.001 pmid: 19723493
51 Miyamoto J, Hasegawa S, Kasubuchi M, Ichimura A, Nakajima A, Kimura I. Nutritional signaling via free fatty acid receptors. Int J Mol Sci 2016; 17(4): 450
https://doi.org/10.3390/ijms17040450 pmid: 27023530
52 Meloni AR, DeYoung MB, Lowe C, Parkes DG. GLP-1 receptor activated insulin secretion from pancreatic β-cells: mechanism and glucose dependence. Diabetes Obes Metab 2013; 15(1): 15–27
https://doi.org/10.1111/j.1463-1326.2012.01663.x pmid: 22776039
53 Insull W Jr. Clinical utility of bile acid sequestrants in the treatment of dyslipidemia: a scientific review. South Med J 2006; 99(3): 257–273
https://doi.org/10.1097/01.smj.0000208120.73327.db pmid: 16553100
54 Sonne DP, Hansen M, Knop FK. Bile acid sequestrants in type 2 diabetes: potential effects on GLP1 secretion. Eur J Endocrinol 2014; 171(2): R47–R65
https://doi.org/10.1530/EJE-14-0154 pmid: 24760535
55 Potthoff MJ, Potts A, He T, Duarte JA, Taussig R, Mangelsdorf DJ, Kliewer SA, Burgess SC. Colesevelam suppresses hepatic glycogenolysis by TGR5-mediated induction of GLP-1 action in DIO mice. Am J Physiol Gastrointest Liver Physiol 2013; 304(4): G371–G380
https://doi.org/10.1152/ajpgi.00400.2012 pmid: 23257920
56 Harach T, Pols TW, Nomura M, Maida A, Watanabe M, Auwerx J, Schoonjans K. TGR5 potentiates GLP-1 secretion in response to anionic exchange resins. Sci Rep 2012; 2(1): 430
https://doi.org/10.1038/srep00430 pmid: 22666533
57 Mazidi M, Rezaie P, Karimi E, Kengne AP. The effects of bile acid sequestrants on lipid profile and blood glucose concentrations: a systematic review and meta-analysis of randomized controlled trials. Int J Cardiol 2017; 227: 850–857
https://doi.org/10.1016/j.ijcard.2016.10.011 pmid: 28029410
58 Hansen M, Sonne DP, Mikkelsen KH, Gluud LL, Vilsbøll T, Knop FK. Bile acid sequestrants for glycemic control in patients with type 2 diabetes: a systematic review with meta-analysis of randomized controlled trials. J Diabetes Complications 2017; 31(5): 918–927
https://doi.org/10.1016/j.jdiacomp.2017.01.011 pmid: 28238556
59 Chen L, Yao X, Young A, McNulty J, Anderson D, Liu Y, Nystrom C, Croom D, Ross S, Collins J, Rajpal D, Hamlet K, Smith C, Gedulin B. Inhibition of apical sodium-dependent bile acid transporter as a novel treatment for diabetes. Am J Physiol Endocrinol Metab 2012; 302(1): E68–E76
https://doi.org/10.1152/ajpendo.00323.2011 pmid: 21934041
60 Ferrebee CB, Dawson PA. Metabolic effects of intestinal absorption and enterohepatic cycling of bile acids. Acta Pharm Sin B 2015; 5(2): 129–134
https://doi.org/10.1016/j.apsb.2015.01.001 pmid: 26579438
61 McGavigan AK, Garibay D, Henseler ZM, Chen J, Bettaieb A, Haj FG, Ley RE, Chouinard ML, Cummings BP. TGR5 contributes to glucoregulatory improvements after vertical sleeve gastrectomy in mice. Gut 2017; 66(2): 226–234
https://doi.org/10.1136/gutjnl-2015-309871 pmid: 26511794
62 Haeusler RA, Astiarraga B, Camastra S, Accili D, Ferrannini E. Human insulin resistance is associated with increased plasma levels of 12α-hydroxylated bile acids. Diabetes 2013; 62(12): 4184–4191
https://doi.org/10.2337/db13-0639 pmid: 23884887
63 Manoogian ENC, Panda S. Circadian rhythms, time-restricted feeding, and healthy aging. Ageing Res Rev 2017; 39: 59–67
https://doi.org/10.1016/j.arr.2016.12.006 pmid: 28017879
64 Froy O. Circadian aspects of energy metabolism and aging. Ageing Res Rev 2013; 12(4): 931–940
https://doi.org/10.1016/j.arr.2013.09.002 pmid: 24075855
65 Adamovich Y, Rousso-Noori L, Zwighaft Z, Neufeld-Cohen A, Golik M, Kraut-Cohen J, Wang M, Han X, Asher G. Circadian clocks and feeding time regulate the oscillations and levels of hepatic triglycerides. Cell Metab 2014; 19(2): 319–330
https://doi.org/10.1016/j.cmet.2013.12.016 pmid: 24506873
66 Kalsbeek A, la Fleur S, Fliers E. Circadian control of glucose metabolism. Mol Metab 2014; 3(4): 372–383
https://doi.org/10.1016/j.molmet.2014.03.002 pmid: 24944897
67 Ribas-Latre A, Eckel-Mahan K. Interdependence of nutrient metabolism and the circadian clock system: importance for metabolic health. Mol Metab 2016; 5(3): 133–152
https://doi.org/10.1016/j.molmet.2015.12.006 pmid: 26977390
68 Le Martelot G, Claudel T, Gatfield D, Schaad O, Kornmann B, Lo Sasso G, Moschetta A, Schibler U. REV-ERBα participates in circadian SREBP signaling and bile acid homeostasis. PLoS Biol 2009; 7(9): e1000181
https://doi.org/10.1371/journal.pbio.1000181 pmid: 19721697
69 Pathak P, Li T, Chiang JY. Retinoic acid-related orphan receptor α regulates diurnal rhythm and fasting induction of sterol 12α-hydroxylase in bile acid synthesis. J Biol Chem 2013; 288(52): 37154–37165
https://doi.org/10.1074/jbc.M113.485987 pmid: 24226095
70 Bass J, Takahashi JS. Circadian integration of metabolism and energetics. Science 2010; 330(6009): 1349–1354
https://doi.org/10.1126/science.1195027 pmid: 21127246
71 Wada E, Koyanagi S, Kusunose N, Akamine T, Masui H, Hashimoto H, Matsunaga N, Ohdo S. Modulation of peroxisome proliferator-activated receptor-α activity by bile acids causes circadian changes in the intestinal expression of Octn1/Slc22a4 in mice. Mol Pharmacol 2015; 87(2): 314–322
https://doi.org/10.1124/mol.114.094979 pmid: 25422143
72 Ferrell JM, Chiang JY. Short-term circadian disruption impairs bile acid and lipid homeostasis in mice. Cell Mol Gastroenterol Hepatol 2015; 1(6):664–677
https://doi.org/DOI: 10.1016/j.jcmgh.2015.08.003 pmid: 26645046
73 Lis CG, Grutsch JF, Wood P, You M, Rich I, Hrushesky WJ. Circadian timing in cancer treatment: the biological foundation for an integrative approach. Integr Cancer Ther 2003; 2(2): 105–111
https://doi.org/10.1177/1534735403002002002 pmid: 15035897
74 Zhang YK, Yeager RL, Klaassen CD. Circadian expression profiles of drug-processing genes and transcription factors in mouse liver. Drug Metab Dispos 2009; 37(1): 106–115
https://doi.org/10.1124/dmd.108.024174 pmid: 18838502
75 Sinal CJ, Yoon M, Gonzalez FJ. Antagonism of the actions of peroxisome proliferator-activated receptor-α by bile acids. J Biol Chem 2001; 276(50): 47154–47162
https://doi.org/10.1074/jbc.M107000200 pmid: 11606578
[1] Hui Wang, Yang Zhang, Zhujun Shen, Ligang Fang, Zhenyu Liu, Shuyang Zhang. Prognostic value of fasting glucose on the risk of heart failure and left ventricular systolic dysfunction in non-diabetic patients with ST-segment elevation myocardial infarction[J]. Front. Med., 2021, 15(1): 70-78.
[2] Di Cheng, Chunyan Hu, Rui Du, Hongyan Qi, Lin Lin, Xueyan Wu, Lina Ma, Kui Peng, Mian Li, Min Xu, Yu Xu, Yufang Bi, Weiqing Wang, Yuhong Chen, Jieli Lu. Serum uric acid and risk of incident diabetes in middle-aged and elderly Chinese adults: prospective cohort study[J]. Front. Med., 2020, 14(6): 802-810.
[3] Huiwen Ren, Can Wu, Ying Shao, Shuang Liu, Yang Zhou, Qiuyue Wang. Correlation between serum miR-154-5p and urinary albumin excretion rates in patients with type 2 diabetes mellitus: a cross-sectional cohort study[J]. Front. Med., 2020, 14(5): 642-650.
[4] Pengju Zhang, Tao Li, Xingyun Wu, Edouard C. Nice, Canhua Huang, Yuanyuan Zhang. Oxidative stress and diabetes: antioxidative strategies[J]. Front. Med., 2020, 14(5): 583-600.
[5] Ning Jiang, Yao Li, Ting Shu, Jing Wang. Cytokines and inflammation in adipogenesis: an updated review[J]. Front. Med., 2019, 13(3): 314-329.
[6] Xiaoqing Li, Xinxin Li, Genbei Wang, Yan Xu, Yuanyuan Wang, Ruijia Hao, Xiaohui Ma. Xiao Ke Qing improves glycometabolism and ameliorates insulin resistance by regulating the PI3K/Akt pathway in KKAy mice[J]. Front. Med., 2018, 12(6): 688-696.
[7] Liping Xuan, Zhiyun Zhao, Xu Jia, Yanan Hou, Tiange Wang, Mian Li, Jieli Lu, Yu Xu, Yuhong Chen, Lu Qi, Weiqing Wang, Yufang Bi, Min Xu. Type 2 diabetes is causally associated with depression: a Mendelian randomization analysis[J]. Front. Med., 2018, 12(6): 678-687.
[8] Ruiting Han, Junli Ma, Houkai Li. Mechanistic and therapeutic advances in non-alcoholic fatty liver disease by targeting the gut microbiota[J]. Front. Med., 2018, 12(6): 645-657.
[9] Jiemin Pan, Weiping Jia. Early-onset diabetes: an epidemic in China[J]. Front. Med., 2018, 12(6): 624-633.
[10] Meng Dong, Jun Lin, Wonchung Lim, Wanzhu Jin, Hyuek Jong Lee. Role of brown adipose tissue in metabolic syndrome, aging, and cancer cachexia[J]. Front. Med., 2018, 12(2): 130-138.
[11] Chao Chen, Chang Wang, Chun Hu, Yachun Han, Li Zhao, Xuejing Zhu, Li Xiao, Lin Sun. Normoalbuminuric diabetic kidney disease[J]. Front. Med., 2017, 11(3): 310-318.
[12] Palka Kaur Khanuja,Satish Chander Narula,Rajesh Rajput,Rajinder Kumar Sharma,Shikha Tewari. Association of periodontal disease with glycemic control in patients with type 2 diabetes in Indian population[J]. Front. Med., 2017, 11(1): 110-119.
[13] Huiqin Zhong,Ya Shao,Ling Fan,Tangshen Zhong,Lu Ren,Yan Wang. Perceived resource support for chronic illnesses among diabetics in north-western China[J]. Front. Med., 2016, 10(2): 219-227.
[14] Juan Zheng,Shih-Lung Woo,Xiang Hu,Rachel Botchlett,Lulu Chen,Yuqing Huo,Chaodong Wu. Metformin and metabolic diseases: a focus on hepatic aspects[J]. Front. Med., 2015, 9(2): 173-186.
[15] Jie Zheng,Yuzhen Gao,Yuejuan Jing,Xiaoshuang Zhou,Yuanyuan Shi,Yanhong Li,Lihua Wang,Ruiying Wang,Maolian Li,Chuanshi Xiao,Yafeng Li,Rongshan Li. Gender differences in the relationship between plasma lipids and fasting plasma glucose in non-diabetic urban Chinese population: a cross-section study[J]. Front. Med., 2014, 8(4): 477-483.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed