Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
FASEB J ; 14(12): 1784-92, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10973928

ABSTRACT

The device of new hepatotrophic prodrugs of the antiviral nucleoside 9-(2-phosphonylmethoxyethyl)adenine (PMEA) with specificity for the asialoglycoprotein receptor on parenchymal liver cells is described. PMEA was conjugated to bi- and trivalent cluster glycosides (K(GN)(2) and K(2)(GN)(3), respectively) with nanomolar affinity for the asialoglycoprotein receptor. The liver uptake of the PMEA prodrugs was more than 10-fold higher than that of the parent drug (52+/-6% and 62+/-3% vs. 4.8+/-0.7% of the injected dose for PMEA) and could be attributed for 90% to parenchymal cells. Accumulation of the PMEA prodrugs in extrahepatic tissue (e.g., kidney, skin) was substantially reduced. The ratio of parenchymal liver cell-to-kidney uptake-a measure of the prodrugs therapeutic window-was increased from 0.058 +/- 0.01 for PMEA to 1.86 +/- 0.57 for K(GN)(2)-PMEA and even 2.69 +/- 0.24 for K(2)(GN)(3)-PMEA. Apparently both glycosides have a similar capacity to redirect (antiviral) drugs to the liver. After cellular uptake, both PMEA prodrugs were converted into the parent drug, PMEA, during acidification of the lysosomal milieu (t(1/2) approximately 100 min), and the released PMEA was rapidly translocated into the cytosol. The antiviral activity of the prodrugs in vitro was dramatically enhanced as compared to the parent drug (5- and 52-fold for K(GN)(2)-PMEA and K(2)(GN)(3)-PMEA, respectively). Given the 15-fold enhanced liver uptake of the prodrugs, we anticipate that the potency in vivo will be similarly increased. We conclude that PMEA prodrugs have been developed with greatly improved pharmacokinetics and therapeutic activity against viral infections that implicate the liver parenchyma (e.g., HBV). In addition, the significance of the above prodrug concept also extends to drugs that intervene in other liver disorders such as cholestasis and dyslipidemia.


Subject(s)
Adenine/analogs & derivatives , Adenine/pharmacology , Antiviral Agents/pharmacology , Herpesvirus 1, Human/drug effects , Liver/metabolism , Organophosphonates , Prodrugs/pharmacology , Adenine/blood , Adenine/metabolism , Adenine/pharmacokinetics , Animals , Antiviral Agents/blood , Antiviral Agents/metabolism , Antiviral Agents/pharmacokinetics , Humans , Male , Prodrugs/metabolism , Prodrugs/pharmacokinetics , Rats , Rats, Wistar , Tritium , Tumor Cells, Cultured
2.
Antimicrob Agents Chemother ; 44(3): 477-83, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10681306

ABSTRACT

Our aim is to selectively deliver 9-(2-phosphonylmethoxyethyl)adenine (PMEA) to parenchymal liver cells, the primary site of hepatitis B virus (HBV) infection. Selective delivery is necessary because PMEA, which is effective against HBV in vitro, is hardly taken up by the liver in vivo. Lactosylated reconstituted high-density lipoprotein (LacNeoHDL), a lipid particle that is specifically internalized by parenchymal liver cells via the asialoglycoprotein receptor, was used as the carrier. PMEA could be incorporated into the lipid moiety of LacNeoHDL by attaching, via an acid-labile bond, lithocholic acid-3alpha-oleate to the drug. The uptake of the lipophilic prodrug (PMEA-LO) by the liver was substantially increased after incorporation into LacNeoHDL. Thirty minutes after injection of [(3)H]PMEA-LO-loaded LacNeoHDL into rats, the liver contained 68.9% +/- 7.7% of the dose (free [(3)H]PMEA, <5%). Concomitantly, the uptake by the kidney was reduced to <2% of the dose (free [(3)H]PMEA, >45%). The hepatic uptake of PMEA-LO-loaded LacNeoHDL occurred mainly by parenchymal cells (88.5% +/- 8.2% of the hepatic uptake). Moreover, asialofetuin inhibited the liver association by >75%, indicating uptake via the asialoglycoprotein receptor. The acid-labile linkage in PMEA-LO, designed to release PMEA during lysosomal processing of the prodrug-loaded carrier, was stable at physiological pH but was hydrolyzed at lysosomal pH (half-life, 60 to 70 min). Finally, subcellular fractionation indicates that the released PMEA is translocated to the cytosol, where it is converted into its active diphosphorylated metabolite. In conclusion, lipophilic modification and incorporation of PMEA into LacNeoHDL improves the biological fate of the drug and may lead to an enhanced therapeutic efficacy against chronic hepatitis B.


Subject(s)
Adenine/analogs & derivatives , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacokinetics , Hepatitis B/drug therapy , Liver/metabolism , Organophosphonates , Adenine/administration & dosage , Adenine/blood , Adenine/chemistry , Adenine/pharmacokinetics , Animals , Antiviral Agents/blood , Antiviral Agents/chemistry , Chromatography/methods , Drug Carriers , Lipoproteins, HDL/metabolism , Lithocholic Acid/analogs & derivatives , Lithocholic Acid/chemical synthesis , Lithocholic Acid/metabolism , Lithocholic Acid/pharmacology , Liver/cytology , Liver/drug effects , Male , Prodrugs/administration & dosage , Prodrugs/chemical synthesis , Prodrugs/pharmacokinetics , Rats , Rats, Wistar , Tissue Distribution , Tritium
3.
Infect Immun ; 66(11): 5107-12, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9784510

ABSTRACT

Lipopolysaccharide (LPS) is cleared from the blood mainly by the liver. The Kupffer cells are primarily responsible for this clearance; liver endothelial and parenchymal cells contribute to a lesser extent. Although several binding sites have been described, only CD14 is known to be involved in LPS signalling. Among the other LPS binding sites that have been identified are scavenger receptors. Scavenger receptor class A (SR-A) types I and II are expressed in the liver on endothelial cells and Kupffer cells, and a 95-kDa receptor, identified as macrosialin, is expressed on Kupffer cells. In this study, we examined the role of scavenger receptors in the binding of LPS by the liver in vivo and in vitro. Fucoidin, a scavenger receptor ligand, significantly reduced the clearance of 125I-LPS from the serum and decreased the liver uptake of 125I-LPS about 40%. Within the liver, the in vivo binding of 125I-LPS to Kupffer and liver endothelial cells was decreased 72 and 71%, respectively, while the binding of 125I-LPS to liver parenchymal cells increased 34% upon fucoidin preinjection. Poly(I) inhibited the binding of 125I-LPS to Kupffer and endothelial cells in vitro 73 and 78%, respectively, while poly(A) had no effect. LPS inhibited the binding of acetylated low-density lipoprotein (acLDL) to Kupffer and liver endothelial cells 40 and 55%, respectively, and the binding of oxidized LDL (oxLDL) to Kupffer and liver endothelial cells 65 and 61%, respectively. oxLDL and acLDL did not significantly inhibit the binding of LPS to these cells. We conclude that on both endothelial cells and Kupffer cells, LPS binds mainly to scavenger receptors, but SR-A and macrosialin contribute to a limited extent to the binding of LPS.


Subject(s)
Endothelium, Vascular/metabolism , Kupffer Cells/metabolism , Lipopolysaccharides/metabolism , Liver/metabolism , Membrane Proteins , Receptors, Immunologic/metabolism , Receptors, Immunologic/physiology , Receptors, Lipoprotein , Animals , Asialoglycoprotein Receptor , Binding Sites/drug effects , Binding, Competitive/drug effects , CD36 Antigens/metabolism , CD36 Antigens/physiology , Endothelium, Vascular/cytology , Humans , Injections, Intravenous , Iodine Radioisotopes , Kupffer Cells/cytology , Ligands , Lipopolysaccharides/pharmacology , Liver/cytology , Male , Organ Specificity , Polysaccharides/administration & dosage , Polysaccharides/pharmacology , Rats , Rats, Wistar , Receptors, Scavenger , Scavenger Receptors, Class A , Scavenger Receptors, Class B
4.
Hepatology ; 26(5): 1303-10, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9362376

ABSTRACT

Recently, we developed a series of cluster mannosides that were able to inhibit tissue-type plasminogen activator (t-PA) binding to the isolated mannose receptor. The mannoside with the highest affinity was able to inhibit t-PA clearance by the liver in the rat. To test whether these mannosides would also be efficient inhibitors in humans, we studied the expression of the mannose receptor in the human liver and determined the efficacy of the mannosides to inhibit mannose receptor-mediated t-PA degradation by both rat and human cells. Immunohistochemistry indicates that, like the rat, human liver endothelial cells and human Kupffer cells do express the mannose receptor. The mannosides do inhibit mannose receptor-mediated t-PA binding, association, and degradation by isolated rat liver endothelial cells and t-PA association and degradation by cultured human macrophages at similar concentrations. The cluster mannoside with six mannose residues connected with a backbone of five lysine groups (M6L5) was, like unlabeled t-PA, able to inhibit 125I-t-PA degradation in the nmol/L range, while the mannoside M5L4 inhibited 125I-t-PA degradation in the micromol/L range. The concentrations of mannoside necessary to inhibit 125I-t-PA degradation in vitro were comparable with the concentrations necessary to inhibit mannose receptor-mediated 125I-t-PA clearance in vivo. We conclude that there is no species difference between rat and humans with respect to the distribution of the mannose receptor in the liver and the affinity of the cluster mannosides, establishing the relevance of the inhibition of mannose receptor-mediated t-PA clearance by M6L5 as observed in the rat, for the human situation.


Subject(s)
Lectins, C-Type , Mannose-Binding Lectins , Mannosides/pharmacology , Receptors, Cell Surface/physiology , Tissue Plasminogen Activator/antagonists & inhibitors , Tissue Plasminogen Activator/metabolism , Animals , Cells, Cultured , Endothelium/cytology , Endothelium/metabolism , Humans , Immunohistochemistry , Kupffer Cells/metabolism , Liver/cytology , Liver/metabolism , Macrophages/metabolism , Male , Mannose Receptor , Proteins/physiology , Rats , Rats, Wistar
5.
Hepatology ; 22(5): 1538-46, 1995 Nov.
Article in English | MEDLINE | ID: mdl-7590673

ABSTRACT

During sepsis the infiltration of leukocytes plays a pivotal role in tissue damage. Induction of septic shock results in an early accumulation of polymorphonuclear leukocytes in the liver (after 3 hours), which is followed by an infiltration of mononuclear phagocytes (after 30 hours). Expression of adhesion molecules may contribute to the migration of leukocytes to the site of inflammation. Therefore, in the present study we determined the expression of intercellular adhesion molecule-1 (ICAM-1) and vascular adhesion molecule-1 (VCAM-1) on hepatocytes, liver endothelial cells, and Kupffer cells after lipopolysaccharide (LPS) treatment of rats in vivo. Parenchymal cells showed no constitutive expression of VCAM-1 and the expression could not be upregulated by LPS treatment in vivo, whereas Kupffer and endothelial cells had a low basal expression of VCAM-1 and this expression was increased 40-fold by LPS treatment in vivo. All three cell types showed a basal expression of ICAM-1 and the expression on endothelial liver cells of untreated rats was two times higher than the expression on parenchymal and Kupffer cells. Stimulation with LPS increased the expression of ICAM-1 2.5 times per parenchymal cells and approximately 4 times for endothelial and Kupffer cells. It is concluded that the expression of adhesion molecules may contribute to the influx of leukocytes during septic shock and, therefore, play a role in tissue damage during septic shock.


Subject(s)
Intercellular Adhesion Molecule-1/biosynthesis , Lipopolysaccharides/pharmacology , Liver/metabolism , Vascular Cell Adhesion Molecule-1/biosynthesis , Animals , Endothelium/metabolism , Endothelium/pathology , Kupffer Cells/metabolism , Kupffer Cells/pathology , Liver/pathology , Male , Rats , Rats, Wistar , Shock, Septic/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...