951
|
Režen T. The impact of cholesterol and its metabolites on drug metabolism. Expert Opin Drug Metab Toxicol 2011; 7:387-98. [PMID: 21320036 DOI: 10.1517/17425255.2011.558083] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Global prevalence of Western-type diet has increased in the last decades resulting in occurrence of certain chronic diseases. This type of diet is also linked to high-cholesterol intake and increase in blood cholesterol. Many of the molecular mechanisms of dealing with increased levels of cholesterol and its metabolites have been elucidated in animal models and humans. It is also evident that cholesterol metabolism is closely connected to drug metabolism. Cholesterol/bile acids and drugs share many transporters, enzymes and regulatory proteins which are key points in the crosstalk. AREAS COVERED This review presents an overview of the effect of cholesterol and its metabolites on drug metabolism with special emphasis on species-specific differences. The article focuses on the role of nuclear receptors farnesoid X receptor, vitamin D receptor and liver X receptor in the regulation of drug metabolism genes and the role of cholesterol biosynthesis intermediates, oxysterols and bile acids in the induction of drug metabolism through pregnane X receptor. EXPERT OPINION Studies show that the regulation of drug metabolism by sterols is multileveled. Many species-dependent differences were observed which hinder the transfer of findings from model animals to humans. As of now, there is little evidence available for cholesterol impact on drug metabolism in vivo in humans. There is also the need to confirm the results obtained in animal models and in vitro analyses in human cells but this is very difficult given the current lack of tools.
Collapse
Affiliation(s)
- Tadeja Režen
- Faculty of Medicine, University of Ljubljana, Institute of Biochemistry, Vrazov Trg 2, SI-1000 Ljubljana, Slovenia.
| |
Collapse
|
952
|
Keitel V, Görg B, Bidmon HJ, Zemtsova I, Spomer L, Zilles K, Häussinger D. The bile acid receptor TGR5 (Gpbar-1) acts as a neurosteroid receptor in brain. Glia 2011; 58:1794-805. [PMID: 20665558 DOI: 10.1002/glia.21049] [Citation(s) in RCA: 196] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
TGR5 (Gpbar-1) is a membrane-bound bile acid receptor in the gastrointestinal tract and immune cells with pleiotropic actions. As shown in the present study, TGR5 is also expressed in astrocytes and neurons. Here, TGR5 may act as a neurosteroid receptor, which is activated by nanomolar concentrations of 5β-pregnan-3α-ol-20-one and micromolar concentrations of 5β-pregnan-3α-17α-21-triol-20-one and 5α-pregnan-3α-ol-20-one (allopregnanolone). TGR5 stimulation in astrocytes and neurons is coupled to adenylate cyclase activation, elevation of intracellular Ca(2+) and the generation of reactive oxygen species. In cultured rat astrocytes, TGR5 mRNA is downregulated in the presence of neurosteroids and ammonia already at concentrations of 0.5 mmol L(-1). Furthermore, TGR5 protein levels are significantly reduced in isolated rat astrocytes after incubation with ammonia. A marked downregulation of TGR5 mRNA is also found in cerebral cortex from cirrhotic patients dying with hepatic encephalopathy (HE) when compared with brains from noncirrhotic control subjects. It is concluded that TGR5 is a novel neurosteroid receptor in brain with implications for the pathogenesis of HE.
Collapse
Affiliation(s)
- Verena Keitel
- Clinic for Gastroenterology, Hepatology, and Infectiology, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | | | | | | | | | | | | |
Collapse
|
953
|
Marion TL, Perry CH, St Claire RL, Yue W, Brouwer KLR. Differential disposition of chenodeoxycholic acid versus taurocholic acid in response to acute troglitazone exposure in rat hepatocytes. Toxicol Sci 2011; 120:371-80. [PMID: 21262925 DOI: 10.1093/toxsci/kfr014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Inhibition of bile acid (BA) transport may contribute to the hepatotoxicity of troglitazone (TRO), a peroxisome proliferator-activated receptor gamma agonist. Typically, studies use taurocholic acid (TCA) as a model substrate to investigate effects of xenobiotics on BA disposition. However, TRO may differentially affect the transport of individual BAs, potentially causing hepatocyte accumulation of more cytotoxic BAs. The effects of TRO on the disposition of [(14)C]-labeled chenodeoxycholic acid ([(14)C]CDCA), an unconjugated cytotoxic BA, were determined in suspended hepatocytes and sandwich-cultured hepatocytes (SCH) from rats. (E)-3-[[[3-[2-(7-chloro-2-quinolinyl)ethenyl]phenyl][[3-(dimethylamino)-3-oxopropyl]thio]methyl]thio]-propanoic acid (MK571), a multidrug resistance-associated protein (MRP) inhibitor, was included to evaluate involvement of MRPs in CDCA disposition. Accumulation in cells + bile of total [(14)C]CDCA species in SCH was sixfold greater than [(3)H]TCA and unaffected by 1 and 10μM TRO; 100μM TRO and 50μM MK571 ablated biliary excretion and significantly increased intracellular accumulation of total [(14)C]CDCA species. Results were similar in Mrp2-deficient TR(-) rat hepatocytes. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis revealed that taurine- and glycine-conjugated CDCA, in addition to unconjugated CDCA, accumulated in hepatocytes during the 10-min incubation. In suspended rat hepatocytes, initial [(14)C]CDCA uptake was primarily Na(+)-independent, whereas initial [(3)H]TCA uptake was primarily Na(+)-dependent; TRO and MK571 decreased [(14)C]CDCA uptake to a lesser extent than [(3)H]TCA. Unexpectedly, MK571 inhibited Na(+)-taurocholate cotransporting polypeptide and bile salt export pump. Differential effects on uptake and efflux of individual BAs may contribute to TRO hepatotoxicity. Although TCA is the prototypic BA used to investigate the effects of xenobiotics on BA transport, it may not be reflective of other BAs.
Collapse
Affiliation(s)
- Tracy L Marion
- Curriculum in Toxicology, UNC School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7270, USA
| | | | | | | | | |
Collapse
|
954
|
Bile acid stimulates hepatocyte polarization through a cAMP-Epac-MEK-LKB1-AMPK pathway. Proc Natl Acad Sci U S A 2011; 108:1403-8. [PMID: 21220320 DOI: 10.1073/pnas.1018376108] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
This study describes a unique function of taurocholate in bile canalicular formation involving signaling through a cAMP-Epac-MEK-Rap1-LKB1-AMPK pathway. In rat hepatocyte sandwich cultures, polarization was manifested by sequential progression of bile canaliculi from small structures to a fully branched network. Taurocholate accelerated canalicular network formation and concomitantly increased cAMP, which were prevented by adenyl cyclase inhibitor. The cAMP-dependent PKA inhibitor did not prevent the taurocholate effect. In contrast, activation of Epac, another cAMP downstream kinase, accelerated canalicular network formation similar to the effect of taurocholate. Inhibition of Epac downstream targets, Rap1 and MEK, blocked the taurocholate effect. Taurocholate rapidly activated MEK, LKB1, and AMPK, which were prevented by inhibition of adenyl cyclase or MEK. Our previous study showed that activated-LKB1 and AMPK participate in canalicular network formation. Linkage between bile acid synthesis, hepatocyte polarization, and regulation of energy metabolism is likely important in normal hepatocyte development and disease.
Collapse
|
955
|
Lasher CD, Rajagopalan P, Murali TM. Discovering networks of perturbed biological processes in hepatocyte cultures. PLoS One 2011; 6:e15247. [PMID: 21245926 PMCID: PMC3016309 DOI: 10.1371/journal.pone.0015247] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Accepted: 11/02/2010] [Indexed: 12/20/2022] Open
Abstract
The liver plays a vital role in glucose homeostasis, the synthesis of bile acids and the detoxification of foreign substances. Liver culture systems are widely used to test adverse effects of drugs and environmental toxicants. The two most prevalent liver culture systems are hepatocyte monolayers (HMs) and collagen sandwiches (CS). Despite their wide use, comprehensive transcriptional programs and interaction networks in these culture systems have not been systematically investigated. We integrated an existing temporal transcriptional dataset for HM and CS cultures of rat hepatocytes with a functional interaction network of rat genes. We aimed to exploit the functional interactions to identify statistically significant linkages between perturbed biological processes. To this end, we developed a novel approach to compute Contextual Biological Process Linkage Networks (CBPLNs). CBPLNs revealed numerous meaningful connections between different biological processes and gene sets, which we were successful in interpreting within the context of liver metabolism. Multiple phenomena captured by CBPLNs at the process level such as regulation, downstream effects, and feedback loops have well described counterparts at the gene and protein level. CBPLNs reveal high-level linkages between pathways and processes, making the identification of important biological trends more tractable than through interactions between individual genes and molecules alone. Our approach may provide a new route to explore, analyze, and understand cellular responses to internal and external cues within the context of the intricate networks of molecular interactions that control cellular behavior.
Collapse
Affiliation(s)
- Christopher D. Lasher
- Genetics, Bioinformatics, and Computational Biology PhD Program, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
| | - Padmavathy Rajagopalan
- Department of Chemical Engineering, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
- ICTAS Center for Systems Biology of Engineered Tissues, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
| | - T. M. Murali
- Department of Computer Science, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
- ICTAS Center for Systems Biology of Engineered Tissues, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
- * E-mail:
| |
Collapse
|
956
|
A contribution to the study of hydrophobicity (lipophilicity) of bile acids with an emphasis on oxo derivatives of 5β-cholanoic acid. HEMIJSKA INDUSTRIJA 2011. [DOI: 10.2298/hemind100924071p] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Due to their promotory action on the transport of some drugs through various
membranes (lipophilic barriers), oxo derivatives of bile acids have recently
been increasingly used in biopharmacy. These compounds exhibit also a lower
membranolytic (toxic) activity than their hydroxy analogues. Because of that
it is of special importance to find out the descriptors that would adequately
describe the structure of bile acids and their biological activity and be
used to model the quantitative structure-activity relationship. In view of
this, the present work is concerned with the application of the
chromatographic parameter RM0 obtained by normal-phase thin-layer
chromatography in the solvent system toluene-butanol and silica gel as
stationary phase to describe the lipophilicity of bile acids. Also, the work
introduces a new molecular descriptor (ND) that reflects both 2D and 3D
topological characteristics of the molecule. Between the retention constant,
RM0 and the descriptor ND there is a good correlation, and both RM0, and ND
describe sufficiently well the structural (conformational) changes that arise
in the process of oxidation of the OH group of the steroid skeleton to an oxo
group. On the other hand, the in silico descriptors of lipophilicity, logP
(atomic-based prediction) and ClogP (fragment-based prediction) predict the
hydrophobicity of bile acid oxo derivatives with a certain error.
Collapse
|
957
|
Tsai SJJ, Zhong YS, Weng JF, Huang HH, Hsieh PY. Determination of bile acids in pig liver, pig kidney and bovine liver by gas chromatography-chemical ionization tandem mass spectrometry with total ion chromatograms and extraction ion chromatograms. J Chromatogr A 2011; 1218:524-33. [DOI: 10.1016/j.chroma.2010.11.062] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 11/19/2010] [Accepted: 11/25/2010] [Indexed: 12/11/2022]
|
958
|
Iguchi Y, Nishimaki-Mogami T, Yamaguchi M, Teraoka F, Kaneko T, Une M. Effects of Chemical Modification of Ursodeoxycholic Acid on TGR5 Activation. Biol Pharm Bull 2011; 34:1-7. [DOI: 10.1248/bpb.34.1] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yusuke Iguchi
- Faculty of Pharmaceutical Science, Hiroshima International University
| | | | | | - Fumiteru Teraoka
- Faculty of Pharmaceutical Science, Hiroshima International University
| | - Tetsuo Kaneko
- Faculty of Pharmaceutical Science, Hiroshima International University
| | - Mizuho Une
- Faculty of Pharmaceutical Science, Hiroshima International University
| |
Collapse
|
959
|
Anakk S, Watanabe M, Ochsner SA, McKenna NJ, Finegold MJ, Moore DD. Combined deletion of Fxr and Shp in mice induces Cyp17a1 and results in juvenile onset cholestasis. J Clin Invest 2011; 121:86-95. [PMID: 21123943 PMCID: PMC3007143 DOI: 10.1172/jci42846] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2010] [Accepted: 10/13/2010] [Indexed: 12/17/2022] Open
Abstract
Bile acid homeostasis is tightly regulated via a feedback loop operated by the nuclear receptors farnesoid X receptor (FXR) and small heterodimer partner (SHP). Contrary to current models, which place FXR upstream of SHP in a linear regulatory pathway, here we show that the phenotypic consequences in mice of the combined loss of both receptors are much more severe than the relatively modest impact of the loss of either Fxr or Shp alone. Fxr-/-Shp-/- mice exhibited cholestasis and liver injury as early as 3 weeks of age, and this was linked to the dysregulation of bile acid homeostatic genes, particularly cytochrome P450, family 7, subfamily a, polypeptide 1 (Cyp7a1). In addition, double-knockout mice showed misregulation of genes in the C21 steroid biosynthesis pathway, with strong induction of cytochrome P450, family 17, subfamily a, polypeptide 1 (Cyp17a1), resulting in elevated serum levels of its enzymatic product 17-hydroxyprogesterone (17-OHP). Treatment of WT mice with 17-OHP was sufficient to induce liver injury that reproduced many of the histopathological features observed in the double-knockout mice. Therefore, our data indicate a pathologic role for increased production of 17-hydroxy steroid metabolites in liver injury and suggest that Fxr-/-Shp-/- mice could provide a model for juvenile onset cholestasis.
Collapse
Affiliation(s)
- Sayeepriyadarshini Anakk
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA.
Department of Internal Medicine, School of Medicine, Keio University, Shinjuku-ku, Tokyo, Japan.
Department of Pathology, Baylor College of Medicine, Houston, Texas, USA
| | - Mitsuhiro Watanabe
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA.
Department of Internal Medicine, School of Medicine, Keio University, Shinjuku-ku, Tokyo, Japan.
Department of Pathology, Baylor College of Medicine, Houston, Texas, USA
| | - Scott A. Ochsner
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA.
Department of Internal Medicine, School of Medicine, Keio University, Shinjuku-ku, Tokyo, Japan.
Department of Pathology, Baylor College of Medicine, Houston, Texas, USA
| | - Neil J. McKenna
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA.
Department of Internal Medicine, School of Medicine, Keio University, Shinjuku-ku, Tokyo, Japan.
Department of Pathology, Baylor College of Medicine, Houston, Texas, USA
| | - Milton J. Finegold
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA.
Department of Internal Medicine, School of Medicine, Keio University, Shinjuku-ku, Tokyo, Japan.
Department of Pathology, Baylor College of Medicine, Houston, Texas, USA
| | - David D. Moore
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA.
Department of Internal Medicine, School of Medicine, Keio University, Shinjuku-ku, Tokyo, Japan.
Department of Pathology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
960
|
Stieger B. The role of the sodium-taurocholate cotransporting polypeptide (NTCP) and of the bile salt export pump (BSEP) in physiology and pathophysiology of bile formation. Handb Exp Pharmacol 2011:205-59. [PMID: 21103971 DOI: 10.1007/978-3-642-14541-4_5] [Citation(s) in RCA: 209] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bile formation is an important function of the liver. Bile salts are a major constituent of bile and are secreted by hepatocytes into bile and delivered into the small intestine, where they assist in fat digestion. In the small intestine, bile salts are almost quantitatively reclaimed and transported back via the portal circulation to the liver. In the liver, hepatocytes take up bile salts and secrete them again into bile for ongoing enterohepatic circulation. Uptake of bile salts into hepatocytes occurs largely in a sodium-dependent manner by the sodium taurocholate cotransporting polypeptide NTCP. The transport properties of NTCP have been extensively characterized. It is an electrogenic member of the solute carrier family of transporters (SLC10A1) and transports predominantly bile salts and sulfated compounds, but is also able to mediate transport of additional substrates, such as thyroid hormones, drugs and toxins. It is highly regulated under physiologic and pathophysiologic conditions. Regulation of NTCP copes with changes of bile salt load to hepatocytes and prevents entry of cytotoxic bile salts during liver disease. Canalicular export of bile salts is mediated by the ATP-binding cassette transporter bile salt export pump BSEP (ABCB11). BSEP constitutes the rate limiting step of hepatocellular bile salt transport and drives enterohepatic circulation of bile salts. It is extensively regulated to keep intracellular bile salt levels low under normal and pathophysiologic situations. Mutations in the BSEP gene lead to severe progressive familial intrahepatic cholestasis. The substrates of BSEP are practically restricted to bile salts and their metabolites. It is, however, subject to inhibition by endogenous metabolites or by drugs. A sustained inhibition will lead to acquired cholestasis, which can end in liver injury.
Collapse
Affiliation(s)
- Bruno Stieger
- Division of Clinical Pharmacology and Toxicology, University Hospital, 8091, Zurich, Switzerland.
| |
Collapse
|
961
|
Detzel CJ, Kim Y, Rajagopalan P. Engineered three-dimensional liver mimics recapitulate critical rat-specific bile acid pathways. Tissue Eng Part A 2010; 17:677-89. [PMID: 20929286 DOI: 10.1089/ten.tea.2010.0423] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
A critical hepatic function is the maintenance of optimal bile acid (BA) compositions to achieve cholesterol homeostasis. BAs are rarely quantified to assess hepatic phenotype in vitro since existing analytical techniques have inadequate resolution. We report a detailed investigation into the biosynthesis and homeostasis of eight primary rat BAs in conventional in vitro hepatocyte cultures and in an engineered liver mimic. The three-dimensional (3D) liver mimic was assembled with layers of primary rat hepatocytes and liver sinusoidal endothelial cells. A high-pressure liquid chromatography and mass spectrometry technique was developed with a detection limit of 1 ng/mL for each BA, which is significantly lower than previous approaches. Over a 2-week culture, only 3D liver mimics exhibited the ratio of conjugated cholic acid to chenodeoxycholic acid that has been observed in vivo. This ratio, an important marker of BA homeostasis, was significantly higher in stable collagen sandwich cultures indicating significant deviation from physiological behavior. The biosynthesis of tauro-β-muricholic acid, a key primary rat BA, doubled only in the engineered liver mimics while decreasing in the other systems. These trends demonstrate that the 3D liver mimics provide a unique platform to study hepatic metabolism.
Collapse
Affiliation(s)
- Christopher J Detzel
- Department of Chemical Engineering, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | | | | |
Collapse
|
962
|
Goldberg AA, Richard VR, Kyryakov P, Bourque SD, Beach A, Burstein MT, Glebov A, Koupaki O, Boukh-Viner T, Gregg C, Juneau M, English AM, Thomas DY, Titorenko VI. Chemical genetic screen identifies lithocholic acid as an anti-aging compound that extends yeast chronological life span in a TOR-independent manner, by modulating housekeeping longevity assurance processes. Aging (Albany NY) 2010; 2:393-414. [PMID: 20622262 PMCID: PMC2933888 DOI: 10.18632/aging.100168] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In
chronologically aging yeast, longevity can be extended by administering a
caloric restriction (CR) diet or some small molecules. These life-extending
interventions target the adaptable target of rapamycin (TOR) and
cAMP/protein kinase A (cAMP/PKA) signaling pathways that are under the
stringent control of calorie availability. We designed a chemical genetic
screen for small molecules that increase the chronological life span of
yeast under CR by targeting lipid metabolism and modulating housekeeping
longevity pathways that regulate longevity irrespective of the number of
available calories. Our screen identifies lithocholic acid (LCA) as one of
such molecules. We reveal two mechanisms underlying
the life-extending effect of LCA in chronologically aging yeast. One
mechanism operates in a calorie availability-independent fashion and
involves the LCA-governed modulation of housekeeping longevity assurance
pathways that do not overlap with the adaptable TOR and cAMP/PKA pathways.
The other mechanism extends yeast longevity under non-CR conditions and
consists in LCA-driven unmasking of the previously unknown anti-aging
potential of PKA. We provide evidence that LCA modulates housekeeping
longevity assurance pathways by suppressing lipid-induced necrosis,
attenuating mitochondrial fragmentation, altering oxidation-reduction
processes in mitochondria, enhancing resistance to oxidative and thermal
stresses, suppressing mitochondria-controlled apoptosis, and enhancing
stability of nuclear and mitochondrial DNA.
Collapse
|
963
|
Prawitt J, Staels B. Bile Acid Sequestrants: Glucose-Lowering Mechanisms. Metab Syndr Relat Disord 2010; 8 Suppl 1:S3-8. [DOI: 10.1089/met.2010.0096] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Janne Prawitt
- University of Lille Nord de France, Lille, France
- INSERM, U1011, Lille, France
- UDSL, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Bart Staels
- University of Lille Nord de France, Lille, France
- INSERM, U1011, Lille, France
- UDSL, Lille, France
- Institut Pasteur de Lille, Lille, France
| |
Collapse
|
964
|
Kim Y, Rajagopalan P. 3D hepatic cultures simultaneously maintain primary hepatocyte and liver sinusoidal endothelial cell phenotypes. PLoS One 2010; 5:e15456. [PMID: 21103392 PMCID: PMC2980491 DOI: 10.1371/journal.pone.0015456] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2010] [Accepted: 09/23/2010] [Indexed: 01/22/2023] Open
Abstract
Developing in vitro engineered hepatic tissues that exhibit stable phenotype is a major challenge in the field of hepatic tissue engineering. However, the rapid dedifferentiation of hepatic parenchymal (hepatocytes) and non-parenchymal (liver sinusoidal endothelial, LSEC) cell types when removed from their natural environment in vivo remains a major obstacle. The primary goal of this study was to demonstrate that hepatic cells cultured in layered architectures could preserve or potentially enhance liver-specific behavior of both cell types. Primary rat hepatocytes and rat LSECs (rLSECs) were cultured in a layered three-dimensional (3D) configuration. The cell layers were separated by a chitosan-hyaluronic acid polyelectrolyte multilayer (PEM), which served to mimic the Space of Disse. Hepatocytes and rLSECs exhibited several key phenotypic characteristics over a twelve day culture period. Immunostaining for the sinusoidal endothelial 1 antibody (SE-1) demonstrated that rLSECs cultured in the 3D hepatic model maintained this unique feature over twelve days. In contrast, rLSECs cultured in monolayers lost their phenotype within three days. The unique stratified structure of the 3D culture resulted in enhanced heterotypic cell-cell interactions, which led to improvements in hepatocyte functions. Albumin production increased three to six fold in the rLSEC-PEM-Hepatocyte cultures. Only rLSEC-PEM-Hepatocyte cultures exhibited increasing CYP1A1/2 and CYP3A activity. Well-defined bile canaliculi were observed only in the rLSEC-PEM-Hepatocyte cultures. Together, these data suggest that rLSEC-PEM-Hepatocyte cultures are highly suitable models to monitor the transformation of toxins in the liver and their transport out of this organ. In summary, these results indicate that the layered rLSEC-PEM-hepatocyte model, which recapitulates key features of hepatic sinusoids, is a potentially powerful medium for obtaining comprehensive knowledge on liver metabolism, detoxification and signaling pathways in vitro.
Collapse
Affiliation(s)
- Yeonhee Kim
- Department of Chemical Engineering, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
| | - Padmavathy Rajagopalan
- Department of Chemical Engineering, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
- ICTAS Center for Systems Biology of Engineered Tissues, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
- * E-mail:
| |
Collapse
|
965
|
Farnesoid X receptor activation improves erectile dysfunction in models of metabolic syndrome and diabetes. Biochim Biophys Acta Mol Basis Dis 2010; 1812:859-66. [PMID: 21056655 DOI: 10.1016/j.bbadis.2010.10.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Revised: 10/22/2010] [Accepted: 10/26/2010] [Indexed: 12/14/2022]
Abstract
The metabolic syndrome (MetS) is an insulin-resistant state characterized by a cluster of cardiovascular risk factors, including abdominal obesity, hyperglycemia, elevated blood pressure and combined dyslipidemia. In this review, we discuss the potential of farnesoid X receptor (FXR) agonists in the treatment of erectile dysfunction (ED), a multifactorial disorder often comorbid with MetS. FXR not only regulates lipid and glucose homeostasis but also influences endothelial function and atherosclerosis, suggesting a regulatory role for this hormone nuclear receptor in the cardiovascular complications associated with the MetS, including ED. MetS induces ED via several mechanisms, and in particular through endothelial dysfunction in penile vessels. In a high-fat diet rabbit model of MetS, a 3-month treatment with the potent and selective FXR agonist INT-747 restores endothelium-dependent relaxation in isolated cavernous tissue, normalizing responsiveness to acetylcholine and to electrical field stimulation. Accordingly, eNOS expression in the penis is greatly up-regulated by INT-747 treatment. Experiments in a rat model of chemically-induced type 1 diabetes further demonstrate that INT-747 treatment preserves erectile function induced by electrical stimulation of the cavernous nerve. These results add a new facet to the pleiotropic activities mediated by FXR, and reveal novel beneficial effects of FXR activation with potential clinical relevance. This article is part of a Special Issue entitled: Translating nuclear receptors from health to disease.
Collapse
|
966
|
|
967
|
Satapathy SK, Sanyal AJ. Novel treatment modalities for nonalcoholic steatohepatitis. Trends Endocrinol Metab 2010; 21:668-675. [PMID: 20880717 DOI: 10.1016/j.tem.2010.08.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Revised: 08/18/2010] [Accepted: 08/18/2010] [Indexed: 02/06/2023]
Abstract
Nonalcoholic steatohepatitis (NASH) is the most common cause of chronic liver disease in the Western world. It can lead to cirrhosis and hepatocellular cancer, and is independently associated with an increased risk of death due to cardiovascular and liver diseases. Over the past 5 years, numerous advances in understanding its pathogenesis have been made, providing a rationale for translation of this information into clinical trials. Although the optimum therapy is still awaited, there is strong evidence to support the use of vitamin E in selected subjects. Many other potential therapeutic options are now available. In this paper, we review the status of both current and emerging therapeutic strategies for patients with NASH.
Collapse
Affiliation(s)
- Sanjaya K Satapathy
- Division of Gastroenterology, Long Island Jewish Medical Center, North Shore University-Long Island Jewish Health System, 270-05 76th Avenue, New Hyde Park, NY 11040, USA
| | | |
Collapse
|
968
|
Wang XX, Jiang T, Shen Y, Caldas Y, Miyazaki-Anzai S, Santamaria H, Urbanek C, Solis N, Scherzer P, Lewis L, Gonzalez FJ, Adorini L, Pruzanski M, Kopp JB, Verlander JW, Levi M. Diabetic nephropathy is accelerated by farnesoid X receptor deficiency and inhibited by farnesoid X receptor activation in a type 1 diabetes model. Diabetes 2010; 59:2916-27. [PMID: 20699418 PMCID: PMC2963551 DOI: 10.2337/db10-0019] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVE The pathogenesis of diabetic nephropathy is complex and involves activation of multiple pathways leading to kidney damage. An important role for altered lipid metabolism via sterol regulatory element binding proteins (SREBPs) has been recently recognized in diabetic kidney disease. Our previous studies have shown that the farnesoid X receptor (FXR), a bile acid-activated nuclear hormone receptor, modulates renal SREBP-1 expression. The purpose of the present study was then to determine if FXR deficiency accelerates type 1 diabetic nephropathy in part by further stimulation of SREBPs and related pathways, and conversely, if a selective FXR agonist can prevent the development of type 1 diabetic nephropathy. RESEARCH DESIGN AND METHODS Insulin deficiency and hyperglycemia were induced with streptozotocin (STZ) in C57BL/6 FXR KO mice. Progress of renal injury was compared with nephropathy-resistant wild-type C57BL/6 mice given STZ. DBA/2J mice with STZ-induced hyperglycemia were treated with the selective FXR agonist INT-747 for 12 weeks. To accelerate disease progression, all mice were placed on the Western diet after hyperglycemia development. RESULTS The present study demonstrates accelerated renal injury in diabetic FXR KO mice. In contrast, treatment with the FXR agonist INT-747 improves renal injury by decreasing proteinuria, glomerulosclerosis, and tubulointerstitial fibrosis, and modulating renal lipid metabolism, macrophage infiltration, and renal expression of SREBPs, profibrotic growth factors, and oxidative stress enzymes in the diabetic DBA/2J strain. CONCLUSIONS Our findings indicate a critical role for FXR in the development of diabetic nephropathy and show that FXR activation prevents nephropathy in type 1 diabetes.
Collapse
MESH Headings
- Animals
- Crosses, Genetic
- DNA Primers
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 1/physiopathology
- Diabetic Nephropathies/pathology
- Diabetic Nephropathies/physiopathology
- Diabetic Nephropathies/prevention & control
- Female
- Foam Cells/pathology
- Kidney/pathology
- Kidney/physiopathology
- Kidney Glomerulus/pathology
- Macrophages/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Mice, Knockout
- Polymerase Chain Reaction
- Receptors, Cytoplasmic and Nuclear/deficiency
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/physiology
Collapse
Affiliation(s)
- Xiaoxin X. Wang
- Department of Medicine, University of Colorado Denver, and the VA Medical Center, Aurora, Colorado
| | - Tao Jiang
- Department of Medicine, University of Colorado Denver, and the VA Medical Center, Aurora, Colorado
| | - Yan Shen
- Department of Medicine, University of Colorado Denver, and the VA Medical Center, Aurora, Colorado
| | - Yupanqui Caldas
- Department of Medicine, University of Colorado Denver, and the VA Medical Center, Aurora, Colorado
| | - Shinobu Miyazaki-Anzai
- Department of Medicine, University of Colorado Denver, and the VA Medical Center, Aurora, Colorado
| | - Hannah Santamaria
- Department of Medicine, University of Colorado Denver, and the VA Medical Center, Aurora, Colorado
| | - Cydney Urbanek
- Department of Medicine, University of Colorado Denver, and the VA Medical Center, Aurora, Colorado
| | - Nathaniel Solis
- Department of Medicine, University of Colorado Denver, and the VA Medical Center, Aurora, Colorado
| | - Pnina Scherzer
- Nephrology and Hypertension Services, Hadassah University Hospital, Jerusalem, Israel
| | - Linda Lewis
- Department of Medicine, University of Colorado Denver, and the VA Medical Center, Aurora, Colorado
| | - Frank J. Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | | | | - Jeffrey B. Kopp
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Jill W. Verlander
- Department of Medicine, Division of Nephrology, Hypertension, and Transplantation, University of Florida, Gainesville, Florida
| | - Moshe Levi
- Department of Medicine, University of Colorado Denver, and the VA Medical Center, Aurora, Colorado
- Corresponding author: Moshe Levi,
| |
Collapse
|
969
|
Poša M, Pilipović A, Lalić M. The influence of NaCl on hydrophobicity of selected, pharmacologically active bile acids expressed with chromatographic retention index and critical micellar concentration. Colloids Surf B Biointerfaces 2010; 81:336-43. [DOI: 10.1016/j.colsurfb.2010.07.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Revised: 07/12/2010] [Accepted: 07/12/2010] [Indexed: 11/26/2022]
|
970
|
Vignozzi L, Morelli A, Filippi S, Comeglio P, Chavalmane AK, Marchetta M, Toce M, Yehiely-Cohen R, Vannelli GB, Adorini L, Maggi M. Farnesoid X receptor activation improves erectile function in animal models of metabolic syndrome and diabetes. J Sex Med 2010; 8:57-77. [PMID: 20955313 DOI: 10.1111/j.1743-6109.2010.02073.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION The farnesoid X receptor (FXR) is critically involved in the regulation of the hepato-biliary system. Recent data suggest a role for FXR in modulating other metabolic pathways and vascular function. AIM To investigate whether long-term administration of the selective FXR agonist INT-747 ameliorates erectile function, we tested it in two animal models of metabolic derangements: a rabbit model of high-fat diet (HFD)-induced metabolic syndrome (MetS) and a rat model of streptozotocin (STZ)-induced type 1 diabetes. METHODS HFD rabbit or STZ rats with or without chronic INT-747 dosing (10 mg/kg/day for 12 weeks). INT-747 addition to rabbit penile smooth muscle cells (rpSMCs). MAIN OUTCOME MEASURE Effects of INT-747 on metabolic features and erectile function in animal models and clarification of mechanism of action in isolated cells. RESULTS INT-747 dosing normalized visceral adiposity and glucose intolerance in HFD rabbits. INT-747 increased penile FXR expression and partially restored endothelial nitric oxide synthase and dimethylarginine dimethylaminohydrolase 1 expression as well as impaired nitric oxide (NO)-dependent relaxation (improved responsiveness to acetylcholine and electrical field stimulation). INT-747 was also effective in regulating NO downstream events, as shown by increased sodium nitroprusside-induced relaxation. Because phosphodiesterase type 5 and protein kinase G (PKG) were unaltered by INT-747, we analyzed the calcium-sensitizing RhoA/ROCK pathway. HFD increased, and INT-747 normalized, RhoA membrane translocation/activation. RhoA/ROCK signaling inhibition by INT-747 was confirmed in rpSMCs by confocal microscopy, MYPT1-phosphorylation, cytoskeleton remodeling, cell migration, and smooth muscle-related genes expression. In STZ rats, FXR penile expression was not altered but was significantly upregulated by INT-747 dosing. In this model, INT-747 improved penile erection induced by electrical stimulation of cavernous nerve and hypersensitivity to intracavernous injection of a ROCK-inhibitor, Y-27632, without improving hyperglycemia. CONCLUSION In HFD rabbits, INT-747 dosing improved glucose sensitivity and MetS-associated erectile dysfunction, via upregulation of NO transmission and inhibition of RhoA/ROCK pathway. In STZ rats, INT-747 restored in vivo penile erection and sensitivity to ROCK inhibition, independently of effects on glycemia.
Collapse
Affiliation(s)
- Linda Vignozzi
- Sexual Medicine and Andrology Unit, Department of Clinical Physiopathology, University of Florence, Florence, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
971
|
Rizzo G, Passeri D, De Franco F, Ciaccioli G, Donadio L, Rizzo G, Orlandi S, Sadeghpour B, Wang XX, Jiang T, Levi M, Pruzanski M, Adorini L. Functional characterization of the semisynthetic bile acid derivative INT-767, a dual farnesoid X receptor and TGR5 agonist. Mol Pharmacol 2010; 78:617-30. [PMID: 20631053 PMCID: PMC2981390 DOI: 10.1124/mol.110.064501] [Citation(s) in RCA: 155] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2010] [Accepted: 07/14/2010] [Indexed: 12/13/2022] Open
Abstract
Two dedicated receptors for bile acids (BAs) have been identified, the nuclear hormone receptor farnesoid X receptor (FXR) and the G protein-coupled receptor TGR5, which represent attractive targets for the treatment of metabolic and chronic liver diseases. Previous work characterized 6α-ethyl-3α,7α-dihydroxy-5β-cholan-24-oic acid (INT-747), a potent and selective FXR agonist, as well as 6α-ethyl-23(S)-methyl-3α,7α,12α-trihydroxy-5β-cholan-24-oic acid (INT-777), a potent and selective TGR5 agonist. Here we characterize 6α-ethyl-3α,7α,23-trihydroxy-24-nor-5β-cholan-23-sulfate sodium salt (INT-767), a novel semisynthetic 23-sulfate derivative of INT-747. INT-767 is a potent agonist for both FXR (mean EC(50), 30 nM by PerkinElmer AlphaScreen assay) and TGR5 (mean EC(50), 630 nM by time resolved-fluorescence resonance energy transfer), the first compound described so far to potently and selectively activate both BA receptors. INT-767 does not show cytotoxic effects in HepG2 cells, does not inhibit cytochrome P450 enzymes, is highly stable to phase I and II enzymatic modifications, and does not inhibit the human ether-a-go-go-related gene potassium channel. In line with its dual activity, INT-767 induces FXR-dependent lipid uptake by adipocytes, with the beneficial effect of shuttling lipids from central hepatic to peripheral fat storage, and promotes TGR5-dependent glucagon-like peptide-1 secretion by enteroendocrine cells, a validated target in the treatment of type 2 diabetes. Moreover, INT-767 treatment markedly decreases cholesterol and triglyceride levels in diabetic db/db mice and in mice rendered diabetic by streptozotocin administration. Collectively, these preclinical results indicate that INT-767 is a safe and effective modulator of FXR and TGR5-dependent pathways, suggesting potential clinical applications in the treatment of liver and metabolic diseases.
Collapse
Affiliation(s)
- Giovanni Rizzo
- Intercept Pharmaceuticals Italia Srl, Via Togliatti, 06073, Corciano, Perugia, Italia.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
972
|
Zhao X, Fritsche J, Wang J, Chen J, Rittig K, Schmitt-Kopplin P, Fritsche A, Häring HU, Schleicher ED, Xu G, Lehmann R. Metabonomic fingerprints of fasting plasma and spot urine reveal human pre-diabetic metabolic traits. Metabolomics 2010; 6:362-374. [PMID: 20676218 PMCID: PMC2899018 DOI: 10.1007/s11306-010-0203-1] [Citation(s) in RCA: 161] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Accepted: 02/17/2010] [Indexed: 12/17/2022]
Abstract
Impaired glucose tolerance (IGT) which precedes overt type 2 diabetes (T2DM) for decades is associated with multiple metabolic alterations in insulin sensitive tissues. In an UPLC-qTOF-mass spectrometry-driven non-targeted metabonomics approach we investigated plasma as well as spot urine of 51 non-diabetic, overnight fasted individuals aiming to separate subjects with IGT from controls thereby identify pathways affected by the pre-diabetic metabolic state. We could clearly demonstrate that normal glucose tolerant (NGT) and IGT subjects clustered in two distinct groups independent of the investigated metabonome. These findings reflect considerable differences in individual metabolite fingerprints, both in plasma and urine. Pre-diabetes associated alterations in fatty acid-, tryptophan-, uric acid-, bile acid-, and lysophosphatidylcholine-metabolism, as well as the TCA cycle were identified. Of note, individuals with IGT also showed decreased levels of gut flora-associated metabolites namely hippuric acid, methylxanthine, methyluric acid, and 3-hydroxyhippuric acid. The findings of our non-targeted UPLC-qTOF-MS metabonomics analysis in plasma and spot urine of individuals with IGT vs NGT offers novel insights into the metabolic alterations occurring in the long, asymptomatic period preceding the manifestation of T2DM thereby giving prospects for new intervention targets. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1007/s11306-010-0203-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xinjie Zhao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, The Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023 China
| | - Jens Fritsche
- Immatics Biotechnologies GmbH, 72076 Tuebingen, Germany
| | - Jiangshan Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, The Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023 China
| | - Jing Chen
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, The Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023 China
| | - Kilian Rittig
- Department of Internal Medicine 4, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Philippe Schmitt-Kopplin
- Institute for Ecological Chemistry, Helmholtz-Zentrum Muenchen—German Research Center for Environmental Health, Ingoldstaedter Landstraße 1, 85764 Neuherberg, Germany
| | - Andreas Fritsche
- Department of Internal Medicine 4, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Hans-Ulrich Häring
- Department of Internal Medicine 4, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Erwin D. Schleicher
- Division of Clinical Chemistry and Pathobiochemistry, Central Laboratory, University Hospital Tuebingen, Otfried-Mueller-Str. 10, 72076 Tuebingen, Germany
| | - Guowang Xu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, The Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023 China
| | - Rainer Lehmann
- Division of Clinical Chemistry and Pathobiochemistry, Central Laboratory, University Hospital Tuebingen, Otfried-Mueller-Str. 10, 72076 Tuebingen, Germany
| |
Collapse
|
973
|
Abstract
The dialogue between gut hormone, bile acids, and the brain plays an important role in energy homeostasis and the onset of Type 2 diabetes mellitus (T2DM). The present review focuses on: (i) bile acid metabolism and the role of bile acids in the regulation of both glucose homeostasis and the control of hypercholesterolemia; (ii) the role of gut hormones in energy homeostasis; and (iii) translation of the pathophysiology of bile acids and gut hormones into clinical practice. Although definitive mechanisms of action of gut hormones and bile acids have not been elucidated completely, these concepts allow us to understand several pharmacological interventions in the treatment of T2DM. Results from further clinical studies with related therapies will help us determine the role of these treatments in the management of energy homeostasis.
Collapse
Affiliation(s)
- Yifei Zhang
- Shanghai Clinical Center for Endocrine and Metabolic Diseases and Division of Endocrine and Metabolic Diseases of E-Institutes of Shanghai Universities, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | | |
Collapse
|
974
|
Dawson PA, Hubbert ML, Rao A. Getting the mOST from OST: Role of organic solute transporter, OSTalpha-OSTbeta, in bile acid and steroid metabolism. BIOCHIMICA ET BIOPHYSICA ACTA 2010; 1801:994-1004. [PMID: 20538072 PMCID: PMC2911127 DOI: 10.1016/j.bbalip.2010.06.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/05/2010] [Revised: 04/29/2010] [Accepted: 06/02/2010] [Indexed: 01/10/2023]
Abstract
The organic solute transporter (OST)(alpha)-OST(beta) is an unusual heteromeric carrier expressed in a variety of tissues including the small intestine, colon, liver, biliary tract, kidney, and adrenal gland. In polarized epithelial cells, OSTalpha-OSTbeta protein is localized on the basolateral membrane and functions in the export or uptake of bile acids and steroids. This article reviews recent results including studies of knockout mouse models that provide new insights to the role of OSTalpha-OSTbeta in the compartmentalization and metabolism of these important lipids.
Collapse
Affiliation(s)
- Paul A Dawson
- Department of Internal Medicine, Section on Gastroenterology, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157, USA.
| | | | | |
Collapse
|
975
|
Eng H, Niosi M, McDonald TS, Wolford A, Chen Y, Simila STM, Bauman JN, Warmus J, Kalgutkar AS. Utility of the carboxylesterase inhibitor bis-para-nitrophenylphosphate (BNPP) in the plasma unbound fraction determination for a hydrolytically unstable amide derivative and agonist of the TGR5 receptor. Xenobiotica 2010; 40:369-80. [PMID: 20297923 DOI: 10.3109/00498251003706598] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The potent, functional agonist of the bile acid Takeda G-protein-coupled receptor 5 (TGR5), (S)-1-(6-fluoro-2-methyl-3,4-dihydroquinolin-1(2H)-yl)-2-(isoquinolin-5-yloxy)ethanone (3), represents a useful tool to probe in vivo TGR5 pharmacology. Rapid degradation of 3 in both rat and mouse plasma, however, hindered the conduct of in vivo pharmacokinetic/pharmacodynamic investigations (including plasma-free fraction (f(u plasma)) determination) in rodent models of pharmacology. Studies were therefore initiated to understand the biochemical basis for plasma instability so that appropriate methodology could be implemented in in vivo pharmacology studies to prevent the breakdown of 3. Compound 3 underwent amide bond cleavage in both rat and mouse plasma with half-lives (T(1/2)) of 39 + or - 7 and 9.9 + or - 0.1 min. bis(p-nitrophenyl) phosphate (BNPP), a specific inhibitor of carboxylesterases, was found to inhibit hydrolytic cleavage in a time- and concentration-dependent manner, which suggested the involvement of carboxylesterases in the metabolism of 3. In contrast with the findings in rodents, 3 was resistant to hydrolytic cleavage in both dog and human plasma. The instability of 3 was also observed in rat and mouse liver microsomes. beta-Nicotinamide adenine dinucleotide phosphate, reduced form (NADPH)-dependent metabolism of 3 occurred more rapidly (T(1/2) approximately 2.22-6.4 min) compared with the metabolic component observed in the absence of the co-factor (T(1/2) approximately 89-130 min). Oxidative metabolism dominated the NADPH-dependent decline of 3, whereas NADPH-independent metabolism of 3 proceeded via simple amide bond hydrolysis. Compound 3 was highly bound (approximately 95%) to both dog and human plasmas. Rat and mouse plasma, pre-treated with BNPP to inhibit carboxylesterases activity, were used to determine the f(u plasma) of 3. A BNPP concentration of 500 microM was determined to be optimal for these studies. Higher BNPP concentrations (1000 microM) appeared to displace 3 from its plasma protein-binding sites in preclinical species and human. Under the conditions of carboxylesterases-inhibited rat and mouse plasma, the level of protein binding displayed by 3 was similar to those observed in dog and human. In conclusion, a novel system has been devised to measure f(u plasma) for a plasma-labile compound. The BNPP methodology can be potentially applied to stabilize hydrolytic cleavage of structurally diverse carboxylesterase substrates in the plasma (and other tissue), thereby allowing the characterization of pharmacology studies on plasma-labile compounds if and when they emerge as hits in exploratory drug-discovery programmes.
Collapse
Affiliation(s)
- H Eng
- Pharmacokinetics, Dynamics and Metabolism, Groton, CT, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
976
|
Cogliati C, Ragona L, D'Onofrio M, Günther U, Whittaker S, Ludwig C, Tomaselli S, Assfalg M, Molinari H. Site-Specific Investigation of the Steady-State Kinetics and Dynamics of the Multistep Binding of Bile Acid Molecules to a Lipid Carrier Protein. Chemistry 2010; 16:11300-10. [DOI: 10.1002/chem.201000498] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
977
|
Herbert MR, Siegel DL, Staszewski L, Cayanan C, Banerjee U, Dhamija S, Anderson J, Fan A, Wang L, Rix P, Shiau AK, Rao TS, Noble SA, Heyman RA, Bischoff E, Guha M, Kabakibi A, Pinkerton AB. Synthesis and SAR of 2-aryl-3-aminomethylquinolines as agonists of the bile acid receptor TGR5. Bioorg Med Chem Lett 2010; 20:5718-21. [PMID: 20801037 DOI: 10.1016/j.bmcl.2010.08.014] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Revised: 07/31/2010] [Accepted: 08/03/2010] [Indexed: 10/19/2022]
Abstract
Optimization of a screening hit from uHTS led to the discovery of TGR5 agonist 32, which was shown to have activity in a rodent model for diabetes.
Collapse
Affiliation(s)
- Mark R Herbert
- Department of Chemistry, Kalypsys, Inc.,10420 Wateridge Circle, San Diego, CA 92121, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
978
|
Smelt AHM. Triglycerides and gallstone formation. Clin Chim Acta 2010; 411:1625-31. [PMID: 20699090 DOI: 10.1016/j.cca.2010.08.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Revised: 08/02/2010] [Accepted: 08/02/2010] [Indexed: 12/16/2022]
Abstract
Changes in bile acid (BA) metabolism and gallbladder function are critical factors in the pathogenesis of gallstones. Patients with hypertriglyceridemia (HTG) - often overweight and insulin resistant - are at risk for gallstone disease. The question arises whether HTG itself contributes to gallstone formation or whether gallstone disease only associates with this disorder. Triglycerides are formed in response to fluxes of non-esterified fatty acids and glucose. Hypertriglyceridemia results from either overproduction of triglycerides by the liver, impaired lipolysis or a combination of both. Hyperinsulinemia, as observed in the insulin resistant state, stimulates very low-density lipoprotein (VLDL)-triglyceride synthesis. Peroxisome proliferator-activated receptors (PPARs), liver X receptors (LXRs), farnesoid X receptor (FXR) and hepatocyte nuclear factor 4α (HNF4α) are the nuclear receptors involved in the regulation of lipogenesis. Microsomal triglyceride transfer protein (MTP) is involved in the production of VLDL and its activation is also under control of transcription factors as FXR and Forkhead box-O1 (FoxO1). Triglyceride and BA metabolism are linked. There is an inverse relationship between bile acid fluxes and pool size and VLDL production and SHP (small heterodimer partner) and FXR are the link between BAs and TG metabolism. BAs are also ligands for FXR and G-protein-coupled receptors, such as TGR5. FXR activation by BAs suppresses the expression of MTP, transcription factor sterol regulatory element binding protein (SREBP)-1c and other lipogenic genes. LXRs stimulate lipogenesis whereas FXRs inhibit the metabolic process. Synthesis of BAs from cholesterol occurs either via the classical pathway (7α-hydroxylation of cholesterol; CYP7A1) or via the alternate pathway (CYP39A1 or CYP7B1). BAs induce FXR, which inhibits CYP7A1 transcription by activation of SHP and inhibition of HNF4α transactivation. Bile composition (supersaturation with cholesterol), gallbladder dysmotility, inflammation, hypersecretion of mucin gel in the gallbladder and slow large intestinal motility and increased intestinal cholesterol absorption may contribute to the pathogenesis of cholesterol gallstones. In HTG patients supersaturated bile may be related to the presence of obesity rather than to HTG itself. Contraction and relaxation of the gallbladder are regulated by neuronal, hormonal and paracrine factors. Postprandial gallbladder emptying is regulated by cholecystokinin (CCK). Poor postprandial gallbladder contraction may be due to the magnitude of the CCK response and to the amount of CCK receptors in the gallbladder smooth muscle cells. In the fasting state gallbladder motility is associated with the intestinal migrating motor complex (MMC) activity and with elevated plasma motilin levels. Fibroblast growth factor (FGF19), produced on arrival of bile acids in the ileum, is also important for gallbladder motility. Gallbladder motility is impaired in HTG patients compared to BMI matched controls. There is evidence that the gallbladder in HTG is less sensitive to CCK and that this sensitivity improves after reversal of high serum TG levels by use of TG lowering agents. In hypertriglyceridemia TG lowering therapy (fibrates or fish-oil) is essential to prevent cardiovascular disease and pancreatitis. Fibrates, however, also increase the risk for cholelithiasis by increasing biliary cholesterol saturation and by reduction of bile acid synthesis. On the other hand fish-oil decreases biliary cholesterol saturation. Fish-oil may increase bile acid synthesis by activation of 7alpha-hydroxylase and may inhibit VLDL production and secretion through activation of nuclear factors and increased apoB degradation. In HTG patients, gallbladder motility improves during bezafibrate as well as during fish-oil therapy. The question remains whether improvement of gallbladder motility and increased lithogenicity of bile by bezafibrate therapy counteract each other or still result in gallstone formation in HTG patients.
Collapse
Affiliation(s)
- A H M Smelt
- Department of General Internal Medicine, Endocrinology and Metabolism, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
979
|
|
980
|
Li T, Owsley E, Matozel M, Hsu P, Novak CM, Chiang JYL. Transgenic expression of cholesterol 7alpha-hydroxylase in the liver prevents high-fat diet-induced obesity and insulin resistance in mice. Hepatology 2010; 52:678-90. [PMID: 20623580 PMCID: PMC3700412 DOI: 10.1002/hep.23721] [Citation(s) in RCA: 192] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
UNLABELLED Cholesterol 7alpha-hydroxylase (CYP7A1) is the rate-limiting enzyme in the bile acid biosynthetic pathway that converts cholesterol into bile acids in the liver. Recent studies have shown that bile acids may play an important role in maintaining lipid, glucose, and energy homeostasis. However, the role of CYP7A1 in the development of obesity and diabetes is currently unclear. In this study, we demonstrated that transgenic mice overexpressing Cyp7a1 in the liver [i.e., Cyp7a1 transgenic (Cyp7a1-tg) mice] were resistant to high-fat diet (HFD)-induced obesity, fatty liver, and insulin resistance. Cyp7a1-tg mice showed increased hepatic cholesterol catabolism and an increased bile acid pool. Cyp7a1-tg mice had increased secretion of hepatic very low density lipoprotein but maintained plasma triglyceride homeostasis. Gene expression analysis showed that the hepatic messenger RNA expression levels of several critical lipogenic and gluconeogenic genes were significantly decreased in HFD-fed Cyp7a1-tg mice. HFD-fed Cyp7a1-tg mice had increased whole body energy expenditure and induction of fatty acid oxidation genes in the brown adipose tissue. CONCLUSION This study shows that Cyp7a1 plays a critical role in maintaining whole body lipid, glucose, and energy homeostasis. The induction of CYP7A1 expression with the expansion of the hydrophobic bile acid pool may be a potential therapeutic strategy for treating metabolic disorders such as fatty liver diseases, obesity, and diabetes in humans.
Collapse
Affiliation(s)
- Tiangang Li
- Department of Integrative Medical Sciences, Northeastern Ohio Universities’ Colleges of Medicine and Pharmacy, Rootstown, OH
| | - Erika Owsley
- Department of Integrative Medical Sciences, Northeastern Ohio Universities’ Colleges of Medicine and Pharmacy, Rootstown, OH
| | - Michelle Matozel
- Department of Integrative Medical Sciences, Northeastern Ohio Universities’ Colleges of Medicine and Pharmacy, Rootstown, OH
| | - Peter Hsu
- Department of Integrative Medical Sciences, Northeastern Ohio Universities’ Colleges of Medicine and Pharmacy, Rootstown, OH
| | - Colleen M. Novak
- Department of Biological Sciences, Kent State University, Kent, OH
| | - John Y. L. Chiang
- Department of Integrative Medical Sciences, Northeastern Ohio Universities’ Colleges of Medicine and Pharmacy, Rootstown, OH
| |
Collapse
|
981
|
Goldberg AA, Kyryakov P, Bourque SD, Titorenko VI. Xenohormetic, hormetic and cytostatic selective forces driving longevity at the ecosystemic level. Aging (Albany NY) 2010; 2:461-70. [PMID: 20693605 PMCID: PMC2954037 DOI: 10.18632/aging.100186] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023]
Abstract
We recently found that lithocholic acid (LCA), a bile acid, extends yeast longevity. Unlike mammals, yeast do not synthesize bile acids. We therefore propose that bile acids released into the environment by mammals may act as interspecies chemical signals providing longevity benefits to yeast and, perhaps, other species within an ecosystem.
Collapse
|
982
|
Cao R, Cronk ZX, Zha W, Sun L, Wang X, Fang Y, Studer E, Zhou H, Pandak WM, Dent P, Gil G, Hylemon PB. Bile acids regulate hepatic gluconeogenic genes and farnesoid X receptor via G(alpha)i-protein-coupled receptors and the AKT pathway. J Lipid Res 2010; 51:2234-44. [PMID: 20305288 PMCID: PMC2903791 DOI: 10.1194/jlr.m004929] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Bile acids are important regulatory molecules that can activate specific nuclear receptors and cell signaling pathways in the liver and gastrointestinal tract. In the current study, the chronic bile fistula (CBF) rat model and primary rat hepatocytes (PRH) were used to study the regulation of gluconeogenic genes phosphoenolpyruvate carboxykinase (PEPCK) and glucose-6-phosphatase (G-6-Pase) and the gene encoding short heterodimeric partner (SHP) by taurocholate (TCA). The intestinal infusion of TCA into the CBF rat rapidly (1h) activated the AKT (approximately 9-fold) and ERK1/2 (3- to 5-fold) signaling pathways, downregulated (approximately 50%, 30 min) the mRNA levels of PEPCK and G-6-Pase, and induced (14-fold in 3 h) SHP mRNA. TCA rapidly ( approximately 50%, 1-2 h) downregulated PEPCK and G-6-Pase mRNA levels in PRH. The downregulation of these genes by TCA was blocked by pretreatment of PRH with pertussis toxin (PTX). In PRH, TCA plus insulin showed a significantly stronger inhibition of glucose secretion/synthesis from lactate and pyruvate than either alone. The induction of SHP mRNA in PRH was strongly blocked by inhibition of PI3 kinase or PKCzeta by specific chemical inhibitors or knockdown of PKCzeta by siRNA encoded by a recombinant lentivirus. Activation of the insulin signaling pathway appears to be linked to the upregulation of farnesoid X receptor functional activity and SHP induction.
Collapse
Affiliation(s)
- Risheng Cao
- Departments of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298
| | - Zhumei Xu Cronk
- Departments of Biochemistry, Virginia Commonwealth University, Richmond, VA 23298
| | - Weibin Zha
- Departments of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298
| | - Lixin Sun
- Departments of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298
| | - Xuan Wang
- Departments of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298
| | - Youwen Fang
- Departments of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298
| | - Elaine Studer
- Departments of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298
| | - Huiping Zhou
- Departments of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298,Departments of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298,McGuire Veterans Affairs Medical Center, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298
| | - William M. Pandak
- Departments of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298,McGuire Veterans Affairs Medical Center, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298
| | - Paul Dent
- Departments of Biochemistry, Virginia Commonwealth University, Richmond, VA 23298
| | - Gregorio Gil
- Departments of Biochemistry, Virginia Commonwealth University, Richmond, VA 23298
| | - Phillip B. Hylemon
- Departments of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298,Departments of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298,McGuire Veterans Affairs Medical Center, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298,To whom correspondence should be addressed. e-mail
| |
Collapse
|
983
|
Yepuru M, Eswaraka J, Kearbey JD, Barrett CM, Raghow S, Veverka KA, Miller DD, Dalton JT, Narayanan R. Estrogen receptor-{beta}-selective ligands alleviate high-fat diet- and ovariectomy-induced obesity in mice. J Biol Chem 2010; 285:31292-303. [PMID: 20657011 DOI: 10.1074/jbc.m110.147850] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Obesity is an epidemic problem affecting millions of people in the Western hemisphere and costs the United States economy more than $200 billion annually. Currently, there are no effective treatments to combat obesity. Recent studies have implicated the constitutive activity of estrogen receptor (ER) β as an important regulator of metabolic diseases. However, the potential of ER-β-selective ligands to offset obesity is not clear. We evaluated the pharmacological effect of ER-β-selective ligands (β-LGNDs) in animal models of high-fat diet- and ovariectomy-induced obesity. Ligand binding, transactivation, and uterotrophic studies with β-LGNDs demonstrated selectivity for ER-β over ER-α. Animals fed a high-fat diet showed a significant increase in body weight, and this weight gain was attenuated by β-LGNDs. High-fat diet-mediated increases in serum cholesterol, leptin, glucose, and fat accumulation in organs were also reduced by β-LGNDs. In addition, MRI scanning indicated that β-LGNDs altered body composition by reducing fat mass and increasing lean body mass. Organ weights and gene expression analyses demonstrated that adipose tissue is the center of action for β-LGNDs, and the reduction in body weight is likely due to increased energy expenditure. In vitro and in vivo mechanistic studies indicated that the anti-obesity effects of β-LGNDs were due to indirect peroxisome proliferator-activated receptor γ antagonistic actions requiring the ligand binding domain of ER-β and through abrogation of the ability of PGC-1 to coactivate peroxisome proliferator-activated receptor γ. In conclusion, these studies indicate that ligand-activated ER-β is a potential therapeutic target to combat obesity and obesity-related metabolic diseases.
Collapse
Affiliation(s)
- Muralimohan Yepuru
- Preclinical Research and Development, GTx Inc, Memphis, Tennessee 38163, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
984
|
Knop FK. Bile-induced secretion of glucagon-like peptide-1: pathophysiological implications in type 2 diabetes? Am J Physiol Endocrinol Metab 2010; 299:E10-3. [PMID: 20424139 DOI: 10.1152/ajpendo.00137.2010] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
During the last decades it has become clear that bile acids not only act as simple fat solubilizers, but additionally represent complex hormonal metabolic integrators. Bile acids activate both nuclear receptors (controlling transcription of genes involved in for example bile acid, cholesterol, and glucose metabolism) and the cell surface G protein-coupled receptor TGR5 (modulating energy expenditure in brown fat and muscle cells). It has been shown that TGR5 is expressed in enteroendocrine L cells, which secrete the potent glucose-lowering incretin hormone glucagon-like peptide-1 (GLP-1). Recently it was shown that bile acid-induced activation of TGR5 results in intestinal secretion of GLP-1 and that enhanced TGR5 signaling improves postprandial glucose tolerance in diet-induced obese mice. This Perspectives article presents these novel findings in the context of prior studies on nutrient-induced GLP-1 secretion and outlines the potential implications of bile acid-induced GLP-1 secretion in physiological, pathophysiological, and pharmacological perspectives.
Collapse
Affiliation(s)
- Filip K Knop
- Department of Internal Medicine F, Gentofte Hospital, University of Copenhagen, Niels Andersens Vej 65, Hellerup, Denmark.
| |
Collapse
|
985
|
Corpechot C, Chrétien Y, Chazouillères O, Poupon R. Demographic, lifestyle, medical and familial factors associated with primary biliary cirrhosis. J Hepatol 2010; 53:162-9. [PMID: 20471130 DOI: 10.1016/j.jhep.2010.02.019] [Citation(s) in RCA: 161] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2009] [Revised: 02/03/2010] [Accepted: 02/03/2010] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Primary biliary cirrhosis (PBC) is believed to result from the interaction of genetic and environmental factors. The controlled studies aiming to assess risk factors for PBC are still limited. Our aim was to identify risk factors and co-morbidities associated with PBC in a large monocentric cohort. METHODS We enrolled 222 patients with PBC and 509 controls matched for age, gender, and residential location. Standardized questionnaire data, including more than 200 questions regarding demographic and anthropometric features, lifestyle, individual and familial medical history, and reproductive history, were prospectively collected and examined. Risk factors with odds ratio (OR) and confidence intervals (CI) were determined using conditional logistic regression analyses. RESULTS Family history of PBC (OR 6.8, 95% CI 2.8-16.4) or autoimmune thyroid disease (AITD) (OR 7.1, 95% CI 3.5-14.5) in first-degree relatives, and individual history of active or passive smoking (OR 3.1, 95% CI 2.0-5.0), recurrent urinary tract infections (UTI) (OR 2.7; 95% CI 2.0-3.7), AITD (OR 7.7, 95% CI 4.8-12.3), Sjögren syndrome (OR 11.9, 95% CI 5.4-26.3), Raynaud syndrome (OR 7.2, 95% CI 4.3-12.1), pruritus during pregnancy (OR 3.9, 95% CI 2.8-5.3), or abortion (OR 2.0, 95% CI 1.6-2.5) were significantly associated with increased risk of PBC, while use of oral contraceptives (OR 0.6; 95% CI 0.5-0.8) was associated with decreased risk. CONCLUSION This study confirms some of the previously reported risk factors for PBC, namely family history of disease and individual history of smoking, UTI, and autoimmune conditions, and further identifies the use of oral contraceptives as a putative protective factor.
Collapse
Affiliation(s)
- Christophe Corpechot
- Service d'Hépatologie, Centre de référence des Maladies Inflammatoires des Voies Biliaires, Hôpital Saint-Antoine, Assistance Publique - Hôpitaux de Paris (APHP), Paris, France.
| | | | | | | |
Collapse
|
986
|
Miyazaki-Anzai S, Levi M, Kratzer A, Ting TC, Lewis LB, Miyazaki M. Farnesoid X receptor activation prevents the development of vascular calcification in ApoE-/- mice with chronic kidney disease. Circ Res 2010; 106:1807-17. [PMID: 20431060 PMCID: PMC2909765 DOI: 10.1161/circresaha.109.212969] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
RATIONALE Vascular calcification is highly associated with cardiovascular morbidity and mortality, especially in patients with chronic kidney disease. The nuclear receptor farnesoid X receptor (FXR) has been implicated in the control of lipid, carbohydrate and bile acid metabolism in several cell types. Although recent studies have shown that FXR is also expressed in vascular smooth muscle cells, its physiological role in vasculature tissue remains obscure. OBJECTIVE Here, we have examined the role of FXR in vascular calcification. METHODS AND RESULTS The FXR gene, a bile acid nuclear receptor, was highly induced during osteogenic differentiation of bovine calcifying vascular cells (CVCs) and in the aorta of apolipoprotein (Apo)E(-/-) mice with chronic kidney disease which are common tissue culture and mouse model, respectively, for aortic calcification. FXR activation by a synthetic FXR agonist, 6alpha-ethyl chenodeoxycholic acid (INT-747) inhibited phosphate induced-mineralization and triglyceride accumulation in CVCs. FXR dominant negative expression augmented mineralization of CVCs and blocked the anticalcific effect of INT-747 whereas VP16FXR that is a constitutively active form reduced mineralization of CVCs. INT-747 treatment also increased phosphorylated c-Jun N-terminal kinase (JNK). SP600125 (specific JNK inhibitor) significantly induced mineralization of CVCs and alkaline phosphatase expression, suggesting that the anticalcific effect of INT-747 is attributable to JNK activation. We also found that INT-747 ameliorates chronic kidney disease induced-vascular calcification in 5/6 nephrectomized ApoE(-/-) mice without affecting the development of atherosclerosis. CONCLUSIONS These observations provide direct evidence that FXR is a key signaling component in regulation of vascular osteogenic differentiation and, thus representing a promising target for the treatment of vascular calcification.
Collapse
Affiliation(s)
- Shinobu Miyazaki-Anzai
- Division of Renal Diseases and Hypertension, Department of Medicine, Denver VA Medical Center and University of Colorado Denver, Denver, Colorado
| | - Moshe Levi
- Division of Renal Diseases and Hypertension, Department of Medicine, Denver VA Medical Center and University of Colorado Denver, Denver, Colorado
| | - Adelheid Kratzer
- Division of Renal Diseases and Hypertension, Department of Medicine, Denver VA Medical Center and University of Colorado Denver, Denver, Colorado
| | - Tabitha C. Ting
- Division of Renal Diseases and Hypertension, Department of Medicine, Denver VA Medical Center and University of Colorado Denver, Denver, Colorado
| | - Linda B. Lewis
- Division of Renal Diseases and Hypertension, Department of Medicine, Denver VA Medical Center and University of Colorado Denver, Denver, Colorado
| | - Makoto Miyazaki
- Division of Renal Diseases and Hypertension, Department of Medicine, Denver VA Medical Center and University of Colorado Denver, Denver, Colorado
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Denver VA Medical Center and University of Colorado Denver, Denver, Colorado
| |
Collapse
|
987
|
Pereira TN, Walsh MJ, Lewindon PJ, Ramm GA. Paediatric cholestatic liver disease: Diagnosis, assessment of disease progression and mechanisms of fibrogenesis. World J Gastrointest Pathophysiol 2010; 1:69-84. [PMID: 21607144 PMCID: PMC3097948 DOI: 10.4291/wjgp.v1.i2.69] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Revised: 03/26/2010] [Accepted: 04/02/2010] [Indexed: 02/06/2023] Open
Abstract
Cholestatic liver disease causes significant morbidity and mortality in children. The diagnosis and management of these diseases can be complicated by an inability to detect early stages of fibrosis and a lack of adequate interventional therapy. There is no single gold standard test that accurately reflects the presence of liver disease, or that can be used to monitor fibrosis progression, particularly in conditions such as cystic fibrosis. This has lead to controversy over how suspected liver disease in children is detected and diagnosed. This review discusses the challenges in using commonly available methods to diagnose hepatic fibrosis and monitor disease progression in children with cholestatic liver disease. In addition, the review examines the mechanisms hypothesised to be involved in the development of hepatic fibrogenesis in paediatric cholestatic liver injury which may ultimately aid in identifying new modalities to assist in both disease detection and therapeutic intervention.
Collapse
|
988
|
Interplay between cholesterol and drug metabolism. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2010; 1814:146-60. [PMID: 20570756 DOI: 10.1016/j.bbapap.2010.05.014] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Revised: 05/17/2010] [Accepted: 05/24/2010] [Indexed: 12/14/2022]
Abstract
Cholesterol biosynthetic and metabolic pathways contain several branching points towards physiologically active molecules, such as coenzyme Q, vitamin D, glucocorticoid and steroid hormones, oxysterols, or bile acids. Sophisticated regulatory mechanisms are involved in maintenance of the homeostasis of not only cholesterol but also other cholesterogenic molecules. In addition to endogenous cues, cholesterol homeostasis needs to accommodate also to exogenous cues that are imported into the body, such as chemicals and medications. Steroid and nuclear receptors together with sterol regulatory element-binding protein (SREBP) mediate the fine tuning of biosynthetic and metabolic routes as well as transports of cholesterol and its derivatives. Similarly, drug/xenobiotic metabolism is the subject to the feedback regulation of cytochrome P450 enzymes and transporters. The regulatory mechanisms that maintain the homeostasis of cholesterogenic molecules and are involved in drug metabolism share similarities. Cholesterol and cholesterogenic compounds (bile acids, glucocorticoids, vitamin D, etc.) regulate the xenosensor signaling in drug-mediated induction of the major drug-metabolizing cytochrome P450 enzymes. The key cellular receptors, pregnane X receptor (PXR), constitutive androstane receptor (CAR), vitamin D receptor (VDR), and glucocorticoid receptor (GR) provide a functional cross-talk between the pathways maintaining cholesterol homeostasis and controlling the expression of drug-metabolizing enzymes. These receptors serve as metabolic sensors, resulting in a coordinate regulation of cholesterogenic compounds metabolism and of the defense against xenobiotic and endobiotic toxicity. Herein we present a comprehensive review of functional interactions between cholesterol homeostasis and drug metabolism involving the main nuclear and steroid receptors.
Collapse
|
989
|
Han S, Li T, Ellis E, Strom S, Chiang JYL. A novel bile acid-activated vitamin D receptor signaling in human hepatocytes. Mol Endocrinol 2010; 24:1151-64. [PMID: 20371703 PMCID: PMC2875805 DOI: 10.1210/me.2009-0482] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2009] [Accepted: 03/03/2010] [Indexed: 12/12/2022] Open
Abstract
Vitamin D receptor (VDR) is activated by natural ligands, 1alpha, 25-dihydroxy-vitamin D(3) [1alpha,25(OH)(2)-D(3)] and lithocholic acid (LCA). Our previous study shows that VDR is expressed in human hepatocytes, and VDR ligands inhibit bile acid synthesis and transcription of the gene encoding cholesterol 7alpha-hydroxylase (CYP7A1). Primary human hepatocytes were used to study LCA and 1alpha,25(OH)(2)-D(3) activation of VDR signaling. Confocal immunofluorescent microscopy imaging and immunoblot analysis showed that LCA and 1alpha, 25(OH)(2)-D(3) induced intracellular translocation of VDR from the cytosol to the nucleus and also plasma membrane where VDR colocalized with caveolin-1. VDR ligands induced tyrosine phosphorylation of c-Src and VDR and their interaction. Inhibition of c-Src abrogated VDR ligand-dependent inhibition of CYP7A1 mRNA expression. Kinase assays showed that VDR ligands specifically activated the c-Raf/MEK1/2/extracellular signal-regulated kinase (ERK) 1/2 pathway, which stimulates serine phosphorylation of VDR and hepatocyte nuclear factor-4alpha, and their interaction. Mammalian two-hybrid assays showed a VDR ligand-dependent interaction of nuclear receptor corepressor-1 and silencing mediator of retinoid and thyroid with VDR/retinoid X receptor-alpha (RXRalpha). Chromatin immunoprecipitation assays revealed that an ERK1/2 inhibitor reversed VDR ligand-induced recruitment of VDR, RXRalpha, and corepressors to human CYP7A1 promoter. In conclusion, VDR ligands activate membrane VDR signaling to activate the MEK1/2/ERK1/2 pathway, which stimulates nuclear VDR/RXRalpha recruitment of corepressors to inhibit CYP7A1 gene transcription in human hepatocytes. This membrane VDR-signaling pathway may be activated by bile acids to inhibit bile acid synthesis as a rapid response to protect hepatocytes from cholestatic liver injury.
Collapse
Affiliation(s)
- Shuxin Han
- Department of Integrative Medical Sciences, Northeastern Ohio University's Colleges of Medicine and Pharmacy, 4209 State Route 44, Rootstown, Ohio 44272, USA
| | | | | | | | | |
Collapse
|
990
|
Mair M, Zollner G, Schneller D, Musteanu M, Fickert P, Gumhold J, Schuster C, Fuchsbichler A, Bilban M, Tauber S, Esterbauer H, Kenner L, Poli V, Blaas L, Kornfeld JW, Casanova E, Mikulits W, Trauner M, Eferl R. Signal transducer and activator of transcription 3 protects from liver injury and fibrosis in a mouse model of sclerosing cholangitis. Gastroenterology 2010; 138:2499-508. [PMID: 20193684 DOI: 10.1053/j.gastro.2010.02.049] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2009] [Revised: 02/04/2010] [Accepted: 02/16/2010] [Indexed: 01/13/2023]
Abstract
BACKGROUND & AIMS Signal transducer and activator of transcription 3 (Stat3) is the main mediator of interleukin-6-type cytokine signaling required for hepatocyte proliferation and hepatoprotection, but its role in sclerosing cholangitis and other cholestatic liver diseases remains unresolved. METHODS We investigated the role of Stat3 in inflammation-induced cholestatic liver injury and used mice lacking the multidrug resistance gene 2 (mdr2(-/-)) as a model for SC. RESULTS We show that conditional inactivation of Stat3 in hepatocytes and cholangiocytes (stat3(Deltahc)) of mdr2(-/-) mice strongly aggravated bile acid-induced liver injury and fibrosis. A similar phenotype was observed in mdr2(-/-) mice lacking interleukin-6 production. Biochemical and molecular characterization suggested that Stat3 exerts hepatoprotective functions in both hepatocytes and cholangiocytes. Loss of Stat3 led to increased expression of tumor necrosis factor alpha, which might reduce the barrier function of bile ducts. Moreover, Stat3-deficient hepatocytes displayed up-regulation of bile acid biosynthesis genes and down-regulation of hepatoprotective epidermal growth factor receptor and insulin-like growth factor 1 signaling pathways. Consistently, stat3(Deltahc) mice were more sensitive to cholic acid-induced liver damage than control mice. CONCLUSIONS Our data suggest that Stat3 prevents cholestasis and liver damage in sclerosing cholangitis via regulation of pivotal functions in hepatocytes and cholangiocytes.
Collapse
Affiliation(s)
- Markus Mair
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
991
|
Musso G, Gambino R, Cassader M. Emerging molecular targets for the treatment of nonalcoholic fatty liver disease. Annu Rev Med 2010; 61:375-92. [PMID: 20059344 DOI: 10.1146/annurev.med.60.101107.134820] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterized by hepatic fat accumulation in the absence of significant ethanol consumption, viral infection, or other specific causes of liver disease. Currently the most common chronic liver disease, affecting 30% of the Western world, NAFLD may progress to cirrhosis and end-stage liver disease and may increase the risk of developing diabetes and cardiovascular disease. Although its pathogenesis is unclear, NAFLD is tightly associated with insulin resistance and the metabolic syndrome. No established treatment exists, and current research is targeting new molecular mechanisms that underlie NAFLD and associated cardiometabolic disorders. This review discusses some of these emerging molecular mechanisms and their therapeutic implications for the treatment of NAFLD: microRNAs, incretin analogs/antagonists, liver-specific thyromimetics, AMP-activated protein kinase activators, and nuclear receptors farnesoid X receptor and pregane X receptor.
Collapse
|
992
|
Abstract
Diabetes is the leading cause of end-stage renal disease in developed countries. In spite of glucose and blood pressure control, for example by use of angiotensin II receptor blockers, diabetic nephropathy still develops and progresses in affected patients and the development of additional protective therapeutic interventions is, therefore, required. Nuclear hormone receptors are transcription factors that regulate carbohydrate metabolism, lipid metabolism, the immune response, and inflammation. These receptors also modulate the development of fibrosis. As a result of their diverse biological effects, nuclear hormone receptors have become major pharmaceutical targets for the treatment of a host of diseases. The increasing prevalence of diabetic nephropathy has led intense investigation into the role that nuclear hormone receptors may have in slowing or preventing the progression of renal disease. This role of nuclear hormone receptors would be associated with improvements in metabolism, the immune response, and inflammation. Eight nuclear receptors have shown a renoprotective effect in the context of diabetic nephropathy. This Review discusses the evidence regarding the beneficial effects of the activation of these receptors in preventing the progression of diabetic nephropathy and describes how the discovery and development of compounds that modulate the activity of nuclear hormone receptors may provide potential additional therapeutic approaches in the management of diabetic nephropathy.
Collapse
|
993
|
Gadaleta RM, van Mil SWC, Oldenburg B, Siersema PD, Klomp LWJ, van Erpecum KJ. Bile acids and their nuclear receptor FXR: Relevance for hepatobiliary and gastrointestinal disease. Biochim Biophys Acta Mol Cell Biol Lipids 2010; 1801:683-92. [PMID: 20399894 DOI: 10.1016/j.bbalip.2010.04.006] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2010] [Revised: 04/03/2010] [Accepted: 04/09/2010] [Indexed: 12/15/2022]
Abstract
The nuclear receptor Farnesoid X Receptor (FXR) critically regulates nascent bile formation and bile acid enterohepatic circulation. Bile acids and FXR play a pivotal role in regulating hepatic inflammation and regeneration as well as in regulating extent of inflammatory responses, barrier function and prevention of bacterial translocation in the intestinal tract. Recent evidence suggests, that the bile acid-FXR interaction is involved in the pathophysiology of a wide range of diseases of the liver, biliary and gastrointestinal tract, such as cholestatic and inflammatory liver diseases and hepatocellular carcinoma, inflammatory bowel disease and inflammation-associated cancer of the colon and esophagus. In this review we discuss current knowledge of the role the bile acid-FXR interaction has in (patho)physiology of the liver, biliary and gastrointestinal tract, and proposed underlying mechanisms, based on in vitro data and experimental animal models. Given the availability of highly potent synthetic FXR agonists, we focus particularly on potential relevance for human disease.
Collapse
Affiliation(s)
- Raffaella M Gadaleta
- Department of Gastroenterology and Hepatology, University Medical Center Utrecht, Utrecht, The Netherlands.
| | | | | | | | | | | |
Collapse
|
994
|
Mellitzer G, Beucher A, Lobstein V, Michel P, Robine S, Kedinger M, Gradwohl G. Loss of enteroendocrine cells in mice alters lipid absorption and glucose homeostasis and impairs postnatal survival. J Clin Invest 2010; 120:1708-21. [PMID: 20364088 DOI: 10.1172/jci40794] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2009] [Accepted: 01/27/2010] [Indexed: 12/22/2022] Open
Abstract
At least 10 enteroendocrine cell types have been identified, and the peptide hormones they secrete have diverse functions that include regulation of glucose homeostasis, food intake, and gastric emptying. Mice lacking individual enteroendocrine hormones, their receptors, or combinations of these have shed light on the role of these hormones in the regulation of energy homeostasis. However, because enteroendocrine hormones have partially overlapping functions, these loss-of-function studies produced only minor phenotypes, and none of the enteroendocrine hormones was shown to be essential for life. To examine the effect of loss of all enteroendocrine cells and hormones on energy homeostasis, we generated mice with intestinal-specific ablation of the proendocrine transcription factor neurogenin 3 (referred to herein as Ngn3Deltaint mice). Ngn3Deltaint mice were deficient for all enteroendocrine cells and hormones, and died with a high frequency during the first week of life. Mutant mice were growth retarded and had yellowish stool suggestive of steatorrhea. Subsequent analyses revealed that Ngn3Deltaint mice had impaired lipid absorption, reduced weight gain, and improved glucose homeostasis. Furthermore, intestinal epithelium of the mutant mice showed an enlarged proliferative crypt compartment and accelerated cell turnover but no changes to goblet and Paneth cell numbers. Enterocytes had shorter microvilli, but the expression of the main brush border enzymes was unaffected. Our data help unravel the role of enteroendocrine cells and hormones in lipid absorption and maintenance of the intestinal epithelium.
Collapse
Affiliation(s)
- Georg Mellitzer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, INSERM U964, CNRS UMR 7104, Université de Strasbourg, Illkirch, France
| | | | | | | | | | | | | |
Collapse
|
995
|
Sheikh Abdul Kadir SH, Miragoli M, Abu-Hayyeh S, Moshkov AV, Xie Q, Keitel V, Nikolaev VO, Williamson C, Gorelik J. Bile acid-induced arrhythmia is mediated by muscarinic M2 receptors in neonatal rat cardiomyocytes. PLoS One 2010; 5:e9689. [PMID: 20300620 PMCID: PMC2837738 DOI: 10.1371/journal.pone.0009689] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2009] [Accepted: 02/05/2010] [Indexed: 12/21/2022] Open
Abstract
Background Intrahepatic cholestasis of pregnancy (ICP) is a common disease affecting up to 5% of pregnancies and which can cause fetal arrhythmia and sudden intrauterine death. We previously demonstrated that bile acid taurocholate (TC), which is raised in the bloodstream of ICP, can acutely alter the rate and rhythm of contraction and induce abnormal calcium destabilization in cultured neonatal rat cardiomyocytes (NRCM). Apart from their hepatic functions bile acids are ubiquitous signalling molecules with diverse systemic effects mediated by either the nuclear receptor FXR or by a recently discovered G-protein coupled receptor TGR5. We aim to investigate the mechanism of bile-acid induced arrhythmogenic effects in an in-vitro model of the fetal heart. Methods and Results Levels of bile acid transporters and nuclear receptor FXR were studied by quantitative real time PCR, western blot and immunostaining, which showed low levels of expression. We did not observe functional involvement of the canonical receptors FXR and TGR5. Instead, we found that TC binds to the muscarinic M2 receptor in NRCM and serves as a partial agonist of this receptor in terms of inhibitory effect on intracellular cAMP and negative chronotropic response. Pharmacological inhibition and siRNA-knockdown of the M2 receptor completely abolished the negative effect of TC on contraction, calcium transient amplitude and synchronisation in NRCM clusters. Conclusion We conclude that in NRCM the TC-induced arrhythmia is mediated by the partial agonism at the M2 receptor. This mechanism might serve as a promising new therapeutic target for fetal arrhythmia.
Collapse
Affiliation(s)
- Siti H. Sheikh Abdul Kadir
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Institute of Reproductive and Developmental Biology, Imperial College London, Hammersmith Campus, London, United Kingdom
- Faculty of Medicine, Universiti Teknologi MARA, Shah Alam, Selangor, Malaysia
| | - Michele Miragoli
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Shadi Abu-Hayyeh
- Institute of Reproductive and Developmental Biology, Imperial College London, Hammersmith Campus, London, United Kingdom
| | - Alexey V. Moshkov
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Qilian Xie
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Heart Centre, First Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Verena Keitel
- Clinic of Gastroenterology, Hepatology and Infectiology, Heinrich-Heine-University, Düsseldorf, Germany
| | | | - Catherine Williamson
- Institute of Reproductive and Developmental Biology, Imperial College London, Hammersmith Campus, London, United Kingdom
| | - Julia Gorelik
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
996
|
Karpen SJ, Trauner M. The new therapeutic frontier--nuclear receptors and the liver. J Hepatol 2010; 52:455-62. [PMID: 20133000 DOI: 10.1016/j.jhep.2009.12.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Revised: 09/20/2009] [Accepted: 10/09/2009] [Indexed: 02/07/2023]
Abstract
A joint EASL/AASLD Monothematic Conference on 'Nuclear Receptors and Liver Disease' was held from February 27th to March 1st, 2009, in Vienna, Austria, to discuss the latest advances at the forefront of basic and clinical nuclear receptor research and its potential implications for liver diseases. This article reports the highlights of the conference and summarizes the main conclusions emphasizing the relevance for clinical and experimental hepatology. The confluence of nuclear receptors as central transcriptional regulators, acting as sensors and adaptors to many of the small molecules present in the intracellular milieu of all the cells of the liver, provides a current framework to address a broader physiological understanding of the liver. The next stage will be the design and testing of safe and effective therapeutics.
Collapse
Affiliation(s)
- Saul J Karpen
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Texas Children's Liver Center, Baylor College of Medicine, Pediatrics/Liver Center, One Baylor Plaza, Houston, TX 77030, USA
| | | |
Collapse
|
997
|
Le Lay J, Kaestner KH. The Fox genes in the liver: from organogenesis to functional integration. Physiol Rev 2010; 90:1-22. [PMID: 20086072 DOI: 10.1152/physrev.00018.2009] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Formation and function of the liver are highly controlled, essential processes. Multiple signaling pathways and transcriptional regulatory networks cooperate in this complex system. The evolutionarily conserved FOX, for Forkhead bOX, class of transcriptional regulators is critical to many aspects of liver development and function. The FOX proteins are small, mostly monomeric DNA binding factors containing the so-called winged helix DNA binding motif that distinguishes them from other classes of transcription factors. We discuss the biochemical and genetic roles of Foxa, Foxl1, Foxm1, and Foxo, as these have been shown to regulate many processes throughout the life of the organ, controlling both formation and function of the liver.
Collapse
Affiliation(s)
- John Le Lay
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104-6145, USA
| | | |
Collapse
|
998
|
Abstract
Bile acids are generated in the liver and are traditionally recognized for their regulatory role in multiple metabolic processes including bile acid homeostasis, nutrient absorption, and cholesterol homeostasis. Recently, bile acids emerged as signaling molecules that, as ligands for the bile acid receptors farnesoid X receptor (FXR) and TGR5, activate and integrate multiple complex signaling pathways involved in lipid and glucose metabolism. Bile acid sequestrants are pharmacologic molecules that bind to bile acids in the intestine resulting in the interruption of bile acid homeostasis and, consequently, reduction in low-density lipoprotein cholesterol levels in hypercholesterolemia. Bile acid sequestrants also reduce glucose levels and improve glycemic control in persons with type 2 diabetes mellitus (T2DM). This article examines the mechanisms by which bile acid-mediated activation of FXR and TGR5 signaling pathways regulate lipid and glucose metabolism and the potential implications for bile acid sequestrant-mediated regulation of lipid and glucose levels in T2DM.
Collapse
Affiliation(s)
- Bart Staels
- Institut Pasteur de Lille, 1 rue Calmette BP245, 59019 Lille cedex, France
| | - Yehuda Handelsman
- Metabolic Institute of America, 18372 Clark Street, #212, Tarzana, CA 91356 USA
| | - Vivian Fonseca
- Tulane University Health Sciences Center, 1430 Tulane Avenue, SL53, New Orleans, LA 70118 USA
| |
Collapse
|
999
|
Synthesis and structure-activity relationships of a series of 3-aryl-4-isoxazolecarboxamides as a new class of TGR5 agonists. Bioorg Med Chem Lett 2010; 20:1363-7. [PMID: 20097073 DOI: 10.1016/j.bmcl.2010.01.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2009] [Revised: 12/24/2009] [Accepted: 01/04/2010] [Indexed: 11/19/2022]
Abstract
A series of 3-aryl-4-isoxazolecarboxamides identified from a high-throughput screening campaign as novel, potent agonists of the human TGR5 G-protein-coupled receptor is described. Many analogues were readily accessible via solution-phase synthesis which resulted in the rapid identification of key structure-activity relationships (SAR), and the discovery of potent exemplars (up to pEC50=9). Details of the SAR and optimization of this series are presented herein.
Collapse
|
1000
|
Evans KA, Budzik BW, Ross SA, Wisnoski DD, Jin J, Rivero RA, Vimal M, Szewczyk GR, Jayawickreme C, Moncol DL, Rimele TJ, Armour SL, Weaver SP, Griffin RJ, Tadepalli SM, Jeune MR, Shearer TW, Chen ZB, Chen L, Anderson DL, Becherer JD, De Los Frailes M, Colilla FJ. Discovery of 3-aryl-4-isoxazolecarboxamides as TGR5 receptor agonists. J Med Chem 2010; 52:7962-5. [PMID: 19902954 DOI: 10.1021/jm901434t] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A series of 3-aryl-4-isoxazolecarboxamides identified from a high-throughput screening campaign as novel, potent small molecule agonists of the human TGR5 G-protein coupled receptor is described. Subsequent optimization resulted in the rapid identification of potent exemplars 6 and 7 which demonstrated improved GLP-1 secretion in vivo via an intracolonic dose coadministered with glucose challenge in a canine model. These novel TGR5 receptor agonists are potentially useful therapeutics for metabolic disorders such as type II diabetes and its associated complications.
Collapse
Affiliation(s)
- Karen A Evans
- Discovery Research, GlaxoSmithKline Pharmaceuticals, Collegeville, PA 19426-0989, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|