Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Lipid Res ; 40(9): 1604-17, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10484607

ABSTRACT

The substrate specificity of the ileal and the hepatic Na(+)/bile acid cotransporters was determined using brush border membrane vesicles and CHO cell lines permanently expressing the Na(+)/bile acid cotransporters from rabbit ileum or rabbit liver. The hepatic transporter showed a remarkably broad specificity for interaction with cholephilic compounds in contrast to the ileal system. The anion transport inhibitor diisothiocyanostilbene disulfonate (DIDS) is a strong inhibitor of the hepatic Na(+)/bile acid cotransporter, but does not show any affinity to its ileal counterpart. Inhibition studies and uptake measurements with about 40 different bile acid analogues differing in the number, position, and stereochemistry of the hydroxyl groups at the steroid nucleus resulted in clear structure;-activity relationships for the ileal and hepatic bile acid transporters. The affinity to the ileal and hepatic Na(+)/bile acid cotransport systems and the uptake rates by cell lines expressing those transporters as well as rabbit ileal brush border membrane vesicles is primarily determined by the substituents on the steroid nucleus. Two hydroxy groups at position 3, 7, or 12 are optimal whereas the presence of three hydroxy groups decreased affinity. Vicinal hydroxy groups at positions 6 and 7 or a shift of the 7-hydroxy group to the 6-position significantly decreased the affinity to the ileal transporter in contrast to the hepatic system. 6-Hydroxylated bile acid derivatives are preferred substrates of the hepatic Na(+)/bile acid cotransporter. Surprisingly, the 3alpha-hydroxy group being present in all natural bile acids is not essential for high affinity interaction with the ileal and the hepatic bile acid transporter. The 3alpha-hydroxy group seems to be necessary for optimal transport of a bile acid across the hepatocyte canalicular membrane. A modification of bile acids at the 3-position therefore conserves the bile acid character thus determining the 3-position of bile acids as the ideal position for drug targeting strategies using bile acid transport pathways.


Subject(s)
Bile Acids and Salts/metabolism , Carrier Proteins/metabolism , Ileum/metabolism , Liver/metabolism , Organic Anion Transporters, Sodium-Dependent , Sodium/metabolism , Symporters , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Animals , Bile Acids and Salts/chemistry , Biological Transport, Active/drug effects , CHO Cells , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cricetinae , DNA, Complementary/genetics , Ileum/drug effects , In Vitro Techniques , Kinetics , Liver/drug effects , Male , Microvilli/drug effects , Microvilli/metabolism , Rabbits , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Taurocholic Acid/metabolism , Tissue Distribution , Transfection
2.
Eur J Biochem ; 249(2): 456-64, 1997 Oct 15.
Article in English | MEDLINE | ID: mdl-9370354

ABSTRACT

For the investigation of the topology of the rabbit ileal Na+/bile-salt-cotransport system, composed of a 93-kDa integral membrane protein and a peripheral 14-kDa bile-acid-binding protein (ILBP), we have synthesized photolabile dimeric bile-salt-transport inhibitors (photoblockers), G1-X-G2, where two bile acid moieties (G1 and G2) are tethered together via a spacer, X, and where one of the two bile acid moieties carries a photoactivatable group. These photoblockers specifically interact with the ileal Na+/bile-salt-cotransport system as demonstrated by a concentration-dependent inhibition of [3H]cholyltaurine uptake by rabbit ileal brush-border membrane vesicles and by inhibition of photolabeling of the 93-kDa and 14-kDa bile-salt-binding proteins by 7,7-azo and 3,3-azo derivatives of cholyltaurine. Ileal bile-salt uptake was specifically inhibited by the photoblockers, which were not taken up themselves by the small intestine as demonstrated by in vivo ileal perfusion. Dependent on the photoblocker used several polypeptides in the molecular-mass range of 14-130 kDa were labeled. The cytoplasmically attached 14-kDa ILBP was significantly labeled only by inhibitors that are photoactivatable in bile acid moiety G1, suggesting that during binding and translocation of a bile-salt molecule by the ileal bile-salt-transport system the steroid nucleus gets access to the cytoplasmic site of the ileal brush-border membrane first. Photoaffinity labeling in the frozen state with the transportable 3,3-azo and 7,7-azo derivatives of cholyltaurine revealed a time-dependent increase in the extent of labeling of the 14-kDa and 93-kDa proteins, suggesting a labeling of these proteins from the cytoplasmic site of the ileal brush-border membrane. By photoaffinity labeling in the frozen state with the various photoblockers time-dependent changes in the extent of photoaffinity labeling of bile-salt-binding proteins were observed, demonstrating the possibility of topological analysis of the rabbit ileal Na+/bile-salt-cotransport system.


Subject(s)
Carrier Proteins/metabolism , Intestinal Mucosa/metabolism , Microvilli/metabolism , Organic Anion Transporters, Sodium-Dependent , Symporters , Affinity Labels , Animals , Bile Acids and Salts/chemistry , Bile Acids and Salts/metabolism , Biological Transport , Carrier Proteins/isolation & purification , Ileum , Kinetics , Male , Molecular Structure , Molecular Weight , Rabbits
3.
Hepatology ; 22(6): 1801-11, 1995 Dec.
Article in English | MEDLINE | ID: mdl-7489992

ABSTRACT

To obtain prodrugs with affinity to liver parenchymal cells, the hepatic 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors HR 780 and lovastatin (syn. mevinolin) were conjugated with the bile acids cholic acid, taurocholic acid, and glycocholic acid. Hepatic uptake and biliary excretion of the coupled drugs were investigated and compared with the noncoupled drugs. Studies were performed with livers of normal Wistar rats, and TR-/GT- Wistar rats with deficient drug excretion. The experiments showed that the parent drug HR 780 was slowly excreted into bile. In contrast, the excretion of the bile acid-conjugated HR 780 derivatives S 3554 (conjugated with cholate), S 3898 (conjugated with glycocholate), and S 4193 (conjugated with taurocholate) was rapid and very efficient in both groups of rat strains. The bile acid-conjugated HMG-CoA reductase inhibitors showed a 10 to 20 times higher affinity for the uptake systems of bile acids than the noncoupled parent drug compounds, and even higher affinities than the bile acids themselves. The cholate conjugate of HR 780 (compound S 3554) was shown to be a noncompetitive inhibitor of taurocholate uptake and a competitive inhibitor of sodium-independent cholate uptake (Ki = 1 mumol/L). Uptake of radiolabeled S 3554 into isolated rat hepatocytes was observed to be rapid, cell specific, saturable, energy dependent, and carrier mediated. However, the carrier for S 3554 uptake was found not to be the cloned Na(+)-dependent taurocholate cotransporting polypeptide Ntcp. Expression of this carrier cRNA in Xenopus laevis oocytes did not stimulate S 3554 uptake.


Subject(s)
ATP-Binding Cassette Transporters , Bile/metabolism , Enzyme Inhibitors/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Liver/enzymology , Liver/metabolism , Animals , Biological Transport , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cholic Acid , Cholic Acids/metabolism , Enzyme Inhibitors/pharmacokinetics , Glycocholic Acid/metabolism , Kinetics , Lovastatin/chemistry , Lovastatin/metabolism , Lovastatin/pharmacokinetics , Male , Oocytes/metabolism , Phalloidine/pharmacology , Prodrugs , Pyridines/chemistry , Pyridines/metabolism , Pyridines/pharmacokinetics , Rats , Rats, Wistar , Structure-Activity Relationship , Taurocholic Acid/metabolism , Xenopus laevis
4.
Biochim Biophys Acta ; 1227(3): 137-54, 1994 Nov 29.
Article in English | MEDLINE | ID: mdl-7986821

ABSTRACT

The target organ for HMG-CoA reductase inhibitors to decrease cholesterol biosynthesis in hypercholesterolemic patients is the liver. Since bile acids undergo an enterohepatic circulation showing a strict organotropism for the liver and the small intestine, the structural elements of an inhibitor for HMG-CoA reductase were combined with those for specific molecular recognition of a bile acid molecule for selective uptake by hepatocytes. Either, the HMG-CoA reductase inhibitors HR 780 and mevinolin were covalently attached to 3 xi-(omega-aminoalkoxy)-7 alpha, 12 alpha-dihydroxy-5 beta-cholan-24-oic acids to obtain bile acid prodrugs, or the side chain of bile acids at C-17 was replaced by 3,5-dihydroxy-heptanoic acid--a structural element essential for inhibition of HMG-CoA reductase--to obtain hybrid bile acid: HMG-CoA reductase inhibitors. The prodrugs could, as expected, not inhibit rat liver HMG-CoA reductase to a significant extent, whereas the hybrid inhibitors showed a stereospecific inhibition of HMG-CoA reductase from rat liver microsomes with an IC50-value of 0.7 microM for the most potent compound S 2467 and 6 microM for its diastereomere S 2468. Uptake measurements with isolated rat hepatocytes and ileal brush-border membrane vesicles from rabbit small intestine revealed a specific interaction of both classes of bile acid-derived HMG-CoA reductase inhibitors with the hepatocyte and ileocyte bile acid uptake systems. Photoaffinity labeling studies using 3-azi- or 7-azi-derivatives of taurocholate with freshly isolated rat hepatocytes or rabbit ileal brush-border membrane vesicles revealed a specific interaction of bile acid derived HMG-CoA reductase inhibitors with the respective putative bile acid transporters in the liver and the ileum demonstrating the bile acid character of these derivatives, both for the prodrugs and the hybrids. Cholesterol biosynthesis in Hep G2 cells was inhibited by the bile acid prodrugs with IC50-values in the range of 68 nM to 600 nM compared to 13 nM for HR 780 and 130 nM for mevinolin. Among the hybrid inhibitors, S 2467 was the most active compound with an IC50-value of 16 microM compared to 55 microM for its diastereomere S 2468. Preliminary in vivo experiments showed an inhibition of hepatic cholesterol biosynthesis after oral dosage only with prodrugs such as S 3554, whereas the hybrid molecules were inactive after oral application.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Bile Acids and Salts/chemistry , Enzyme Inhibitors/chemical synthesis , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Liver/metabolism , Lovastatin/chemistry , Prodrugs/chemistry , Pyridines/chemistry , Animals , Bile Acids and Salts/metabolism , In Vitro Techniques , Intestine, Small/metabolism , Liver/drug effects , Liver/enzymology , Lovastatin/pharmacology , Male , Microsomes, Liver/drug effects , Prodrugs/metabolism , Pyridines/pharmacokinetics , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Rats, Wistar , Stereoisomerism
5.
J Med Chem ; 37(20): 3240-6, 1994 Sep 30.
Article in English | MEDLINE | ID: mdl-7932551

ABSTRACT

To increase hepatoselectivity of HMG-CoA reductase inhibitors by using the specific bile acid transport systems, deoxycholic acid-derived inhibitors 9 and 11 have been synthesized, on the basis of the concept of combining in one molecule structural requirements for specific inhibition of the HMG-CoA reductase and specific recognition by the ileal bile acid transport system. The 1-methyl-3-carboxylpropyl subunit of deoxycholic acid was replaced by the 3,5-dihydroxyheptanoic acid lactone of lovastatin, and position 12-OH was esterified with 2-methylbutyric acid. Compounds 9 and 11 were evaluated for their inhibitory activity on rat liver HMG-CoA reductase, cholesterol biosynthesis in HEP G2 cells, and [3H]taurocholate uptake in rabbit brush border membrane vesicles and compared with methyl derivatives 8 and 10. The steroidal 21-CH3 group affects both activity on HMG-CoA reductase and recognition by the ileal bile acid transport system.


Subject(s)
Androstanols/chemical synthesis , Bile Acids and Salts/chemistry , Bile Acids and Salts/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Ileum/metabolism , Pyrones/chemical synthesis , Androstanols/pharmacology , Animals , Biological Transport/drug effects , Cell Line , Cholesterol/biosynthesis , Humans , Ileum/drug effects , Male , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Microvilli/drug effects , Microvilli/metabolism , Pyrones/pharmacology , Rabbits , Rats , Rats, Wistar , Sodium/pharmacology , Structure-Activity Relationship , Taurocholic Acid/metabolism , Tritium
7.
J Biol Chem ; 267(26): 18598-604, 1992 Sep 15.
Article in English | MEDLINE | ID: mdl-1526993

ABSTRACT

Bile acids are selectively taken up from portal blood into the liver by specific transport systems in the hepatocyte plasma membrane. Therefore, studies were performed to evaluate the potential of bile acids as shuttles to deliver drugs specifically to the liver. The alkylating cytostatic drug chlorambucil and the fluorescent prolyl-4-hydroxylase inhibitor 4-nitrobenzo-2-oxa-1,3-diazol-beta-Ala-Phe-5-oxaproline-Gly were covalently linked via an amide bond to 7 alpha, 12 alpha,-dihydroxy-3 beta- (omega-aminoalkoxy)-5-beta-cholan-24-oic acid. The chlorambucil-bile acid conjugates S 2521, S 2539, S 2567, and S 2576 inhibited Na(+)-dependent [3H]taurocholate uptake in a concentration-dependent manner both into isolated rat hepatocytes and rabbit ileal brush border membrane vesicles, whereas the parent drug chlorambucil showed no significant inhibitory effect. The chlorambucil-bile acid conjugates were able to prevent photoaffinity labeling of bile acid binding proteins in rat hepatocytes by the photolabile [3H]7,7-azo derivative of taurocholic acid indicating their bile acid character. The chlorambucil-bile acid conjugate S 2577 was able to alkylate proteins demonstrating the drug character conserved in the hybrid-molecules. Liver perfusion experiments revealed a secretion profile of the chlorambucil-bile acid conjugate S 2576 into bile very similar to taurocholate compared to chlorambucil which is predominantly excreted by the kidney. 4-Nitrobenzo-2-oxa-1,3-diazol-beta-Ala-Phe-5-oxaproline-Gly- t-butylester (S 4404), a fluorescent peptide inhibitor of prolyl-4-hydroxylase, was not transported in intact form from portal blood into bile in contrast to its bile acid conjugate S 3744; about 25% of the peptide-bile acid conjugate S 3744 was secreted in intact form into bile within 40 min compared with less than 4% of the parent oxaprolylpeptide S 4404. In conclusion, these studies reveal that modified bile acid molecules can be used as "Trojan horses" to deliver a drug molecule specifically into the liver and the biliary system. This offers important pharmacological options for the development of liver-specific drugs.


Subject(s)
Bile Acids and Salts/metabolism , Chlorambucil/pharmacokinetics , Liver/metabolism , Affinity Labels , Animals , Cells, Cultured , Chlorambucil/metabolism , Chromatography, Thin Layer , Fluorescent Dyes , Ileum/metabolism , Liver/cytology , Male , Microvilli/metabolism , Oligopeptides/metabolism , Oligopeptides/pharmacokinetics , Photochemistry , Rabbits , Rats , Rats, Inbred Strains , Taurocholic Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...