8
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Systemic ASBT inactivation protects against liver damage in obstructive cholestasis in mice

      research-article
      1 , 2 , 1 , 2 , 1 , 2 , 1 , 2 , 1 , 2 , 1 , 2 , 2 , 3 , 1 , 2 , 4 , 1 , 2 , 4 , 1 , 2 , 4 ,
      JHEP Reports
      Elsevier
      Apical sodium-dependent bile acid transporter (ASBT), IBAT, NTCP, BSEP, PFIC, Alagille, Cholestasis, Bile salt pool size, Renal excretion, ALT, alanine transaminase, ASBT, apical sodium-dependent bile acid transporter, ASBTi, ASBT inhibitors, AST, aspartate transaminase, BDL, bile duct ligation, CCl4, carbon tetrachloride, CK7, cytokeratin 7, FRET, Förster resonance energy transfer, G-OCA, glycine-conjugated OCA, HepG2 cell, hepatocarcinoma cell, MDR2, multidrug resistance protein 2, NASH, non-alcoholic steatohepatitis, NGM282, non-tumorigenic fibroblast growth factor 19 analogue, NTCP, Na+/taurocholate cotransporting polypeptide, NucleoBAS, nuclear Bile Acid Sensor, OCA, obeticholic acid, PBC, primary biliary cholangitis, PentaOH, pentahydroxylated, RT-qPCR, real-time quantitative PCR, TCA, taurocholic acid, TetraOH, tetrahydroxylated, T-OCA, taurine-conjugated OCA, U2OS, osteosarcoma cell, UHPLC-MS, ultrahigh-performance liquid chromatography mass spectrometry, WT, wild-type

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background & Aims

          Non-absorbable inhibitors of the apical sodium-dependent bile acid transporter (ASBT; also called ileal bile acid transporter [IBAT]) are recently approved or in clinical development for multiple cholestatic liver disorders and lead to a reduction in pruritus and (markers for) liver injury. Unfortunately, non-absorbable ASBT inhibitors (ASBTi) can induce diarrhoea or may be ineffective if cholestasis is extensive and largely precludes intestinal excretion of bile acids. Systemically acting ASBTi that divert bile salts towards renal excretion may alleviate these issues.

          Methods

          Bile duct ligation (BDL) was performed in ASBT-deficient (ASBT knockout [KO]) mice as a model for chronic systemic ASBT inhibition in obstructive cholestasis. Co-infusion of radiolabelled taurocholate and inulin was used to quantify renal bile salt excretion after BDL. In a second (wild-type) mouse model, a combination of obeticholic acid (OCA) and intestine-restricted ASBT inhibition was used to lower the bile salt pool size before BDL.

          Results

          After BDL, ASBT KO mice had reduced plasma bilirubin and alkaline phosphatase compared with wild-type mice with BDL and showed a marked reduction in liver necrotic areas at histopathological analysis, suggesting decreased BDL-induced liver damage. Furthermore, ASBT KO mice had reduced bile salt pool size, lower plasma taurine-conjugated polyhydroxylated bile salt, and increased urinary bile salt excretion. Pretreatment with OCA + ASBTi in wild-type mice reduced the pool size and greatly improved liver injury markers and liver histology.

          Conclusions

          A reduced bile salt pool at the onset of cholestasis effectively lowers cholestatic liver injury in mice. Systemic ASBT inhibition may be valuable as treatment for cholestatic liver disease by lowering the pool size and increasing renal bile salt output even under conditions of minimal faecal bile salt secretion.

          Lay summary

          Novel treatment approaches against cholestatic liver disease (resulting in reduced or blocked flow of bile) involve non-absorbable inhibitors of the bile acid transport protein ASBT, but these are not always effective and/or can cause unwanted side effects. In this study, we demonstrate that systemic inhibition/inactivation of ASBT protects mice against developing severe cholestatic liver injury after bile duct ligation, by reducing bile salt pool size and increasing renal bile salt excretion.

          Graphical abstract

          Highlights

          • ASBT deficiency in mice markedly reduced bile duct ligation-induced liver injury.

          • Systemic ASBT inactivation lowered total bile salt pool size.

          • Systemic ASBT inactivation increased renal bile salt excretion.

          • Reduced bile salt pool size before cholestasis onset improved liver injury.

          Related collections

          Most cited references25

          • Record: found
          • Abstract: found
          • Article: not found

          Assumption-free analysis of quantitative real-time polymerase chain reaction (PCR) data.

          Quantification of mRNAs using real-time polymerase chain reaction (PCR) by monitoring the product formation with the fluorescent dye SYBR Green I is being extensively used in neurosciences, developmental biology, and medical diagnostics. Most PCR data analysis procedures assume that the PCR efficiency for the amplicon of interest is constant or even, in the case of the comparative C(t) method, equal to 2. The latter method already leads to a 4-fold error when the PCR efficiencies vary over just a 0.04 range. PCR efficiencies of amplicons are usually calculated from standard curves based on either known RNA inputs or on dilution series of a reference cDNA sample. In this paper we show that the first approach can lead to PCR efficiencies that vary over a 0.2 range, whereas the second approach may be off by 0.26. Therefore, we propose linear regression on the Log(fluorescence) per cycle number data as an assumption-free method to calculate starting concentrations of mRNAs and PCR efficiencies for each sample. A computer program to perform this calculation is available on request (e-mail: bioinfo@amc.uva.nl; subject: LinRegPCR).
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Targeted deletion of the ileal bile acid transporter eliminates enterohepatic cycling of bile acids in mice.

            The ileal apical sodium bile acid cotransporter participates in the enterohepatic circulation of bile acids. In patients with primary bile acid malabsorption, mutations in the ileal bile acid transporter gene (Slc10a2) lead to congenital diarrhea, steatorrhea, and reduced plasma cholesterol levels. To elucidate the quantitative role of Slc10a2 in intestinal bile acid absorption, the Slc10a2 gene was disrupted by homologous recombination in mice. Animals heterozygous (Slc10a2+/-) and homozygous (Slc10a2-/-) for this mutation were physically indistinguishable from wild type mice. In the Slc10a2-/- mice, fecal bile acid excretion was elevated 10- to 20-fold and was not further increased by feeding a bile acid binding resin. Despite increased bile acid synthesis, the bile acid pool size was decreased by 80% and selectively enriched in cholic acid in the Slc10a2-/- mice. On a low fat diet, the Slc10a2-/- mice did not have steatorrhea. Fecal neutral sterol excretion was increased only 3-fold, and intestinal cholesterol absorption was reduced only 20%, indicating that the smaller cholic acid-enriched bile acid pool was sufficient to facilitate intestinal lipid absorption. Liver cholesteryl ester content was reduced by 50% in Slc10a2-/- mice, and unexpectedly plasma high density lipoprotein cholesterol levels were slightly elevated. These data indicate that Slc10a2 is essential for efficient intestinal absorption of bile acids and that alternative absorptive mechanisms are unable to compensate for loss of Slc10a2 function.
              Bookmark
              • Record: found
              • Abstract: not found
              • Article: not found

              Effect of ileal bile acid transporter inhibitor GSK2330672 on pruritus in primary biliary cholangitis: a double-blind, randomised, placebo-controlled, crossover, phase 2a study

                Bookmark

                Author and article information

                Contributors
                Journal
                JHEP Rep
                JHEP Rep
                JHEP Reports
                Elsevier
                2589-5559
                27 August 2022
                November 2022
                27 August 2022
                : 4
                : 11
                : 100573
                Affiliations
                [1 ]Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
                [2 ]Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Centers, The Netherlands
                [3 ]Department of Pathology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
                [4 ]Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
                Author notes
                []Corresponding author. Address: Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Meibergdreef 69-71, 1105 BK Amsterdam, The Netherlands. Tel.: +31-020-5668832; Fax: +31-020-5669190. k.f.vandegraaf@ 123456amsterdamumc.nl
                Article
                S2589-5559(22)00145-8 100573
                10.1016/j.jhepr.2022.100573
                9494276
                36160754
                c44721e2-4ea6-4065-ba07-1ef090672f68
                © 2022 The Author(s)

                This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).

                History
                : 16 July 2021
                : 29 July 2022
                : 16 August 2022
                Categories
                Research Article

                apical sodium-dependent bile acid transporter (asbt),ibat,ntcp,bsep,pfic,alagille,cholestasis,bile salt pool size,renal excretion,alt, alanine transaminase,asbt, apical sodium-dependent bile acid transporter,asbti, asbt inhibitors,ast, aspartate transaminase,bdl, bile duct ligation,ccl4, carbon tetrachloride,ck7, cytokeratin 7,fret, förster resonance energy transfer,g-oca, glycine-conjugated oca,hepg2 cell, hepatocarcinoma cell,mdr2, multidrug resistance protein 2,nash, non-alcoholic steatohepatitis,ngm282, non-tumorigenic fibroblast growth factor 19 analogue,ntcp, na+/taurocholate cotransporting polypeptide,nucleobas, nuclear bile acid sensor,oca, obeticholic acid,pbc, primary biliary cholangitis,pentaoh, pentahydroxylated,rt-qpcr, real-time quantitative pcr,tca, taurocholic acid,tetraoh, tetrahydroxylated,t-oca, taurine-conjugated oca,u2os, osteosarcoma cell,uhplc-ms, ultrahigh-performance liquid chromatography mass spectrometry,wt, wild-type

                Comments

                Comment on this article