Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Publication year range
1.
Diabetes Obes Metab ; 18(1): 6-15, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26228188

ABSTRACT

Diabetes mellitus is a growing worldwide epidemic disease, currently affecting 1 in 12 adults. Treatment of disease complications typically consumes ∼10% of healthcare budgets in developed societies. Whilst immune-mediated destruction of insulin-secreting pancreatic ß cells is responsible for Type 1 diabetes, both the loss and dysfunction of these cells underly the more prevalent Type 2 diabetes. The establishment of robust drug development programmes aimed at ß-cell restoration is still hampered by the absence of means to measure ß-cell mass prospectively in vivo, an approach which would provide new opportunities for understanding disease mechanisms and ultimately assigning personalized treatments. In the present review, we describe the progress towards this goal achieved by the Innovative Medicines Initiative in Diabetes, a collaborative public-private consortium supported by the European Commission and by dedicated resources of pharmaceutical companies. We compare several of the available imaging methods and molecular targets and provide suggestions as to the likeliest to lead to tractable approaches. Furthermore, we discuss the simultaneous development of animal models that can be used to measure subtle changes in ß-cell mass, a prerequisite for validating the clinical potential of the different imaging tracers.


Subject(s)
Diabetes Mellitus/pathology , Insulin-Secreting Cells/pathology , Molecular Imaging/methods , Adult , Animals , Cell Adhesion , Glucagon-Like Peptide-1 Receptor/metabolism , Humans , Insulin-Secreting Cells/metabolism , Luminescent Measurements , Manganese , Membrane Glycoproteins/metabolism , Mice , Rats , Sulfonylurea Receptors/metabolism , Vesicular Monoamine Transport Proteins/metabolism , Zinc
2.
Diabetes Obes Metab ; 15(4): 324-34, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23121319

ABSTRACT

AIMS: Intensive glycaemic control in type 2 diabetes achieved by insulin is generally accompanied by body weight gain. This study was performed to emphasize the meaning of caloric analysis of urine and faeces for energy balance. METHODS: We measured energetic loss via urine and faeces during antihyperglycaemic treatment in male obese Zucker diabetic fatty (ZDF) rats. Rats were treated for 10 days with the sodium-glucose-linked transporter-2 (SGLT2) inhibitor AVE2268, with insulin glargine, with the GLP-1 receptor agonist lixisenatide and with the combination of insulin glargine and lixisenatide. Each study was accompanied by one lean (Fa/?) and one obese (fa/fa) untreated non-diabetic and diabetic control group, respectively. Blood glucose, body weight alterations and food assimilation efficiency were monitored. RESULTS: In control ZDF rats, more than 12 g/day of pure glucose was urinarily excreted. In total, the energetic loss via urine exceeded 30% from total energy uptake. Insulin glargine treatment decreased urinary energetic loss, leading to a body weight gain of approximately 3 g/day. An almost body weight-neutral antihyperglycaemic treatment could be achieved with AVE2268 and lixisenatide. While lixisenatide reduced body weight gain via reduction of energy uptake, the SGLT2 inhibitor even increased urinary glucose and thus energy excretion. Combining insulin glargine with lixisenatide attenuated the anabolic effect of insulin resulting in weight neutrality. CONCLUSIONS: Our data clearly show renal contribution to the body's energy control by urinary glucose excretion (UGE) during antidiabetic treatment. The undesired retained energy could be reduced via additional UGE or via simultaneous reduction of energy uptake and/or energy retention.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Glucosides/pharmacology , Hypoglycemic Agents/pharmacology , Insulin, Long-Acting/pharmacology , Peptides/pharmacology , Animals , Blood Glucose/drug effects , Glucagon-Like Peptide-1 Receptor , Glycated Hemoglobin/drug effects , Insulin Glargine , Kidney/drug effects , Male , Rats , Rats, Zucker , Receptors, Glucagon/agonists , Sodium-Glucose Transporter 2 , Sodium-Glucose Transporter 2 Inhibitors , Weight Gain/drug effects
3.
J Biol Chem ; 276(38): 36020-7, 2001 Sep 21.
Article in English | MEDLINE | ID: mdl-11447228

ABSTRACT

Reabsorption of bile acids occurs in the terminal ileum by a Na(+)-dependent transport system composed of several subunits of the ileal bile acid transporter (IBAT) and the ileal lipid-binding protein. To identify the bile acid-binding site of the transporter protein IBAT, ileal brush border membrane vesicles from rabbit ileum were photoaffinity labeled with a radioactive 7-azi-derivative of cholyltaurine followed by enrichment of IBAT protein by preparative SDS gel electrophoresis. Enzymatic fragmentation with chymotrypsin yielded IBAT peptide fragments in the molecular range of 20.4-4 kDa. With epitope-specific antibodies generated against the C terminus a peptide of molecular mass of 6.6-7 kDa was identified as the smallest peptide fragment carrying both the C terminus and the covalently attached radiolabeled bile acid derivative. This clearly indicates that the ileal Na(+)/bile acid cotransporting protein IBAT contains a bile acid-binding site within the C-terminal 56-67 amino acids. Based on the seven-transmembrane domain model for IBAT, the bile acid-binding site is localized to a region containing the seventh transmembrane domain and the cytoplasmic C terminus. Alternatively, assuming the nine-transmembrane domain model, this bile acid-binding site is localized to the ninth transmembrane domain and the C terminus.


Subject(s)
Carrier Proteins/metabolism , Ileum/metabolism , Organic Anion Transporters, Sodium-Dependent , Symporters , Amino Acid Sequence , Animals , Azo Compounds/chemistry , Bile Acids and Salts/metabolism , Binding Sites , Carrier Proteins/chemistry , Chymotrypsin/metabolism , Electrophoresis, Polyacrylamide Gel , In Vitro Techniques , Ligands , Molecular Sequence Data , Photoaffinity Labels , Rabbits
4.
FEBS Lett ; 487(2): 293-7, 2000 Dec 29.
Article in English | MEDLINE | ID: mdl-11150527

ABSTRACT

To identify protein components of the intestinal cholesterol transporter, rabbit small intestinal brush border membrane vesicles were submitted to photoaffinity labeling using photoreactive derivatives of 2-azetidinone cholesterol absorption inhibitors. An integral membrane protein of M(r) 145.3+/-7.5 kDa was specifically labeled in brush border membrane vesicles from rabbit jejunum and ileum. Its labeling was concentration-dependently inhibited by the presence of cholesterol absorption inhibitors whereas bile acids, D-glucose, fatty acids or cephalexin had no effect. The inhibitory potency of 2-azetidinones to inhibit photolabeling of the 145 kDa protein correlated with their in vivo activity to inhibit intestinal cholesterol absorption. These results suggest that an integral membrane protein of M(r) 145 kDa is (a component of) the cholesterol absorption system in the brush border membrane of small intestinal enterocytes.


Subject(s)
Anticholesteremic Agents/pharmacology , Azetidines/pharmacology , Carrier Proteins/metabolism , Cholesterol/analogs & derivatives , Cholesterol/pharmacokinetics , Intestinal Absorption/drug effects , Intestinal Mucosa/physiology , Intestine, Small/physiology , Microvilli/physiology , Animals , Bile Acids and Salts/pharmacology , Cephalexin/pharmacology , Drug Design , Ezetimibe , Fatty Acids/pharmacology , Glucose/pharmacology , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred Strains , Rabbits , Structure-Activity Relationship
5.
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
6.
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
8.
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
...