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










Publication year range
1.
Cardiovasc Drugs Ther ; 27(2): 117-24, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23314760

ABSTRACT

PURPOSE: Although erythropoietin (EPO) is known to express angiogenic and cardioprotective effects, it also induces hypertension, polycythemia, and platelet activation, which may cause serious adverse effects in patients with cardiovascular diseases. We compared the angiogenic effects of EPO and its nonerythropoietic derivative, asialo-EPO (AEPO). METHODS: Lower limb ischemia was induced in ICR and C57/BL mice. Mice were injected intramuscularly with 2 µg/kg of EPO derivatives for 6 or 7 days. To assess biological differences, the tissue affinity of both EPO derivatives was analyzed in vitro using heparin affinity column chromatography. Tissue affinity was also analyzed in vivo using an intramuscular pharmacokinetic study. RESULTS: The survival of ischemic legs was better in the AEPO group than that in the EPO group (5/13 = 38.5 % vs 1/13 = 7.7 %, p < 0.05), and an increase in regenerated vessels was observed in the AEPO group, but not in the EPO group in ICR mice. Vessel/muscle ratios in control, EPO, and AEPO groups were 0.50 ± 0.34, 0.61 ± 0.32, and 2.83 ± 1.13, respectively (p < 0.0001). On the other hand, regenerated vessels were observed in both EPO and AEPO groups (p < 0.001) in C57/BL mice. AEPO, but not EPO, expressed heparin affinity in vitro. Intramuscularly injected EPO gradually decreased in muscle tissue, while AEPO was maintained at 2.5 ng/muscle for 1 day after several hours of a rapid clearance phase in vivo. CONCLUSIONS: AEPO exerts stronger angiogenic effects than those of EPO presumably via its tissue affinity. Administration of AEPO is a promising option for the treatment of patients with critical limb ischemia.


Subject(s)
Asialoglycoproteins/administration & dosage , Erythropoietin/analogs & derivatives , Ischemia/drug therapy , Animals , Asialoglycoproteins/pharmacokinetics , Erythropoietin/administration & dosage , Erythropoietin/pharmacokinetics , Heparin/metabolism , Injections, Intramuscular , Ischemia/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Neovascularization, Physiologic/drug effects , Protein Binding
2.
J Control Release ; 160(1): 81-7, 2012 May 30.
Article in English | MEDLINE | ID: mdl-22342472

ABSTRACT

Cerebral ischemia-reperfusion (I/R) injury induces secondary cerebral damage. As drugs for treating this type of injury have shown poor efficacy and adverse side effects in clinical trials, a novel therapeutic strategy has been long awaited. In this study, we focused on the disruption of the blood-brain barrier after stroke, and applied a liposomal drug delivery system (DDS) designed to enhance the pharmacological effect of the neuroprotectant and to avoid side effects. PEGylated liposomes were injected at varying time after the start of reperfusion in transient middle cerebral artery occlusion (t-MCAO) model rats. The results showed PEGylated liposomes accumulated in the ischemic hemisphere at an early stage after reperfusion and were retained in the lesion for at least 24h after injection. We also investigated the effectiveness of asialo-erythropoietin (AEPO)-modified PEGylated liposomes (AEPO-liposomes) in treating the cerebral I/R injury. AEPO-liposome treatment significantly reduced TTC-defined cerebral legion following cerebral I/R injury, and ameliorated motor function compared with vehicle and AEPO treatment. In conclusion, these results indicate that AEPO-liposomes are a promising liposomal formulation for protecting the brain from I/R injury, and that this liposomal DDS has potential as a novel strategy for the treatment of cerebral I/R injury.


Subject(s)
Asialoglycoproteins/administration & dosage , Drug Delivery Systems/methods , Erythropoietin/analogs & derivatives , Ischemic Attack, Transient/drug therapy , Neuroprotective Agents/administration & dosage , Reperfusion Injury/prevention & control , Animals , Apoptosis/drug effects , Asialoglycoproteins/adverse effects , Asialoglycoproteins/pharmacokinetics , Asialoglycoproteins/therapeutic use , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Cell Culture Techniques , Cell Survival/drug effects , Disease Models, Animal , Erythropoietin/administration & dosage , Erythropoietin/adverse effects , Erythropoietin/pharmacokinetics , Erythropoietin/therapeutic use , In Situ Nick-End Labeling , Ischemic Attack, Transient/complications , Ischemic Attack, Transient/metabolism , Liposomes , Male , Neuroprotective Agents/adverse effects , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/therapeutic use , PC12 Cells , Rats , Rats, Wistar , Reperfusion Injury/etiology , Reperfusion Injury/metabolism , Tissue Distribution
3.
Thromb Haemost ; 104(2): 243-51, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20508904

ABSTRACT

Clearance mechanisms for recombinant activated human FVII (rFVIIa; NovoSeven), a heterogeneously glycosylated protein, have yet to be fully elucidated, but may involve the liver. The effects of the gamma-carboxy glutamic acid (Gla) domain and the sialic acid content of the protein on rFVIIa clearance were investigated following intravenous administration of rFVIIa lacking the Gla domain, des(1-44) rFVIIa and asialo-rFVIIa in pharmacokinetic (PK) studies and perfused rat livers. PK parameters for both rFVIIa and des(1-44) rFVIIa had similar biphasic clearance profiles, as well as half-lives ([t(1/2)]=80 and 88 minutes, respectively), while asialo-rFVIIa was cleared quickly (t(1/2)=21 minutes) with a linear clearance profile. Perfused liver studies with all proteins (10 nM) mirrored the trends in profiles observed in the PK study. rFVIIa and des(1-44) rFVIIa were cleared to a similar extent, 41% and 35%, respectively, after 1 h, whereas plasma-derived FVII from humans (which has a higher sialylation content than rFVIIa) was cleared to a lesser extent (21%). Asialo-rFVIIa, on the other hand, was almost totally cleared and when an excess of asialo-orosomucoid was added to the perfusate, its clearance was significantly reduced (by 34%) and also for rFVIIa, albeit to a lesser extent (by 14%). Together these data suggest that carbohydrate receptor(s) (e.g. the asialoglycoprotein receptor, ASGPR) play a role in asialo-rFVIIa and rFVIIa clearance. In vivo and liver clearance data correlated well showing similar trends and indicated that rFVIIa clearance is not affected by the Gla domain, but rather by a subpopulation of N-glycosylated structures on rFVIIa.


Subject(s)
Asialoglycoproteins/pharmacokinetics , Coagulants/pharmacokinetics , Factor VIIa/pharmacokinetics , Liver/metabolism , Peptide Fragments/pharmacokinetics , Animals , Asialoglycoprotein Receptor/metabolism , Asialoglycoproteins/administration & dosage , Asialoglycoproteins/blood , Asialoglycoproteins/pharmacology , Coagulants/administration & dosage , Coagulants/blood , Factor VIIa/administration & dosage , Glycosylation , Half-Life , Humans , Injections, Intravenous , Liver/drug effects , Male , Metabolic Clearance Rate , Orosomucoid/analogs & derivatives , Orosomucoid/pharmacology , Peptide Fragments/administration & dosage , Peptide Fragments/blood , Perfusion , Rats , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage , Recombinant Proteins/blood , Recombinant Proteins/pharmacokinetics
4.
Drug Metab Pharmacokinet ; 25(1): 101-7, 2010.
Article in English | MEDLINE | ID: mdl-20208393

ABSTRACT

Human alpha(1)-acid glycoprotein (AGP), a serum glycoprotein, is thought to have anti-inflammatory effects by a mechanism that is not well understood. In this study, we investigated the pharmacokinetics of AGP in mice using enzymatically modified AGP (AGP with the sialic acids removed, asialo-AGP, and with both sialic acids and galactose removed, agalacto-AGP). It was observed that AGP was eliminated from the circulation slowly, and was mainly taken up by the liver. The elimination of labeled AGP, asialo-AGP and agalacto-AGP from the circulation was suppressed in the presence of excess unlabeled AGP, asialo-AGP and agalacto-AGP, respectively, suggesting the receptor-mediated uptake of these AGPs. Interestingly, the uptake of AGP by the liver competed with agalacto-AGP, but not with asialo-AGP, while agalacto-AGP competed with asialo-AGP. These results suggest that agalacto-AGP binds to at least two types of receptors, namely asialoglycoprotein receptor (ASGPR) and an as yet unidentified receptor that is shared with AGP, and that AGP is directly taken up by the liver through such a receptor and not via ASGPR. These findings help improve our understanding of the clearance mechanism of AGP.


Subject(s)
Liver/metabolism , Orosomucoid/pharmacokinetics , Animals , Asialoglycoprotein Receptor/drug effects , Asialoglycoproteins/chemical synthesis , Asialoglycoproteins/pharmacokinetics , Binding, Competitive/drug effects , Cells, Cultured , Male , Mice , Mice, Inbred Strains , Orosomucoid/analogs & derivatives , Orosomucoid/chemical synthesis , Tissue Distribution
6.
Nat Med ; 14(6): 648-55, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18488037

ABSTRACT

The Ashwell receptor, the major lectin of hepatocytes, rapidly clears from blood circulation glycoproteins bearing glycan ligands that include galactose and N-acetylgalactosamine. This asialoglycoprotein receptor activity remains a key factor in the development and administration of glycoprotein pharmaceuticals, yet a biological purpose of the Ashwell receptor has remained elusive. We have identified endogenous ligands of the Ashwell receptor as glycoproteins and regulatory components in blood coagulation and thrombosis that include von Willebrand factor (vWF) and platelets. The Ashwell receptor normally modulates vWF homeostasis and is responsible for thrombocytopenia during systemic Streptococcus pneumoniae infection by eliminating platelets desialylated by the bacterium's neuraminidase. Hemostatic adaptation by the Ashwell receptor moderates the onset and severity of disseminated intravascular coagulation during sepsis and improves the probability of host survival.


Subject(s)
Asialoglycoprotein Receptor/blood , Blood Coagulation Disorders/blood , Blood Coagulation Disorders/pathology , Animals , Asialoglycoprotein Receptor/genetics , Asialoglycoprotein Receptor/metabolism , Asialoglycoproteins/blood , Asialoglycoproteins/pharmacokinetics , Blood Platelets/metabolism , Blood Platelets/microbiology , Blood Platelets/pathology , Disease Models, Animal , Fluorescent Antibody Technique, Indirect , Hemostasis/physiology , Hepatocytes/chemistry , Hepatocytes/metabolism , Homozygote , Humans , Ligands , Metabolic Clearance Rate/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Pneumococcal Infections/metabolism , Pneumococcal Infections/pathology , Protein Binding , Thrombocytopenia/microbiology , Thrombocytopenia/pathology , von Willebrand Factor/metabolism
7.
J Dig Dis ; 8(2): 89-95, 2007 May.
Article in English | MEDLINE | ID: mdl-17532821

ABSTRACT

OBJECTIVE: To testify that the asialoorosomucoid (ASOR) prepared by us has liver-targeting specificity and to investigate its pharmacokinetic characteristics. METHODS: The distribution of 125I-ASOR in vivo was determined by single photon emission computed tomography (SPECT) and immunohistochemical technique after 125I-ASOR was injected into Sprague-Dawley (S-D) rats through their caudal veins. In vitro, different doses of pEGFP-N1 plasmid were transfected into both HepG2 cells and HT1080 cells with the use of ASOR-poly-L-lysine. At 24 and 48 h after transfection, the expression of green fluorescent protein (GFP) was determined under fluorescent microscope. Pharmacokinetic parameters were calculated according to two-compartment open system model with first-order kinetics. RESULTS: SPECT images showed that 125I-ASOR was located only in liver/stomach and root of caudal vein/bladder at 10 min after injection. The 125I-ASOR radioactivities of organs taken out from S-D rats were different at different times, and about 63% of 125I-ASOR was located in the liver at 10 min after injection. At 30 min after injection a peak of radioactivity was seen in stomach. The times of these two radioactivity peaks were different. Immunohistochemical study of liver frozen sections showed that ASOR was combined mainly with hepatocyte membrane, especially in areas with rich blood flow. In vitro study showed that ASOR targeted specifically cells with asialoglycoprotein receptor (ASGr). GFP expression was detected in HepG2 cells but not in HT1080 cells. Furthermore, the more quantity of pEGFP-N1 transfected and the longer expression time, the higher GFP expression level was in HepG2 cells. The 125I-ASOR pharmacokinetics equation for liver was Ct=662216e-3.362t+8896e-2343t. 125I-ASOR was excreted from liver slowly after an initial rapid decrease. The pharmacokinetic equation for stomach was Ct=-114815e-1.7t+1148153e-15t and the half-life of 125I-ASOR in stomach was 4.62 h. CONCLUSIONS: ASOR prepared by us could be an efficient gene transfer vector, ASOR was distributed mainly in the liver and stomach and had high targeting specificity to hepatocytes or hepatic originating cells.


Subject(s)
Asialoglycoprotein Receptor/drug effects , Asialoglycoproteins/pharmacokinetics , Glycoproteins/pharmacokinetics , Liver/metabolism , Orosomucoid/analogs & derivatives , Stomach/drug effects , Animals , Asialoglycoproteins/chemistry , Gene Transfer Techniques , Genetic Vectors , Glycoproteins/chemistry , Hepatocytes/metabolism , Injections, Intravenous , Iodine Radioisotopes , Orosomucoid/chemistry , Orosomucoid/pharmacokinetics , Rats , Rats, Sprague-Dawley , Tomography, Emission-Computed, Single-Photon
8.
Glycobiology ; 16(10): 926-37, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16774910

ABSTRACT

Viscumin of mistletoe (Viscum album L.) has a concentration-dependent activity profile unique to plant AB-toxins. It starts with lectin-dependent mitogenicity and then covers toxicity and cell agglutination, associated with shifts in the monomer/dimer equilibrium. Each lectin subunit harbors two sections for ligand contact. In the dimer, the B-chain sites in subdomain 2 gamma (designated as the Tyr-sites) appear fully accessible, whereas Trp-sites in subdomain 1 alpha are close to the dimer interface. It is unclear whether both types of sites operate similarly in binding glycoligands in solution. By systematically covering a broad range of lactose/lectin ratio in isothermal titration calorimetry, we obtained evidence for two sites showing dissimilar binding affinity. Intriguingly, the site with higher affinity was only partially occupied. To assign the observed properties to the Trp/Tyr-sites, we next performed chemically induced dynamic nuclear polarization measurements of Trp and Tyr accessibility. A Tyr signal, but not distinct Trp peaks, was recorded when testing the dimer. Lactose-quenchable Trp peaks became visible on the destabilization of the dimer by citraconylation, intimating Trp involvement in ligand contact in the monomer. Fittingly, Tyr acetylation but not mild Trp oxidation reduced the dimer hemagglutination activity and the extent of binding to asialofetuin-Sepharose 4B. Altogether, the results attribute lectin activity in the dimer primarily to Tyr-sites. Full access to Trp-sites is gained on dimer dissociation. Thus, the monomer/dimer equilibrium of viscumin regulates the operativity of these sites. Their structural divergence affords the possibility for differences in ligand selection when comparing monomers (Tyr- and Trp-sites) with dimers (primarily Tyr-sites).


Subject(s)
Galactosides/metabolism , Lectins/chemistry , Lectins/metabolism , Mistletoe/chemistry , Asialoglycoproteins/pharmacokinetics , Chromatography, Gel/methods , Dimerization , Fetuins , Hemagglutination/drug effects , Hydrogen-Ion Concentration , Lactose/metabolism , Microscopy, Polarization/methods , Mistletoe/metabolism , Models, Molecular , Plant Lectins/chemistry , Plant Lectins/metabolism , Plant Preparations/metabolism , Plant Proteins/metabolism , Protein Binding , Ribosome Inactivating Proteins , Ribosome Inactivating Proteins, Type 2 , Sepharose/pharmacokinetics , Substrate Specificity , Toxins, Biological/metabolism , Tryptophan/chemistry , Tryptophan/metabolism , Tyrosine/chemistry , Tyrosine/metabolism , alpha-Fetoproteins/pharmacokinetics
9.
J Neurochem ; 91(4): 900-10, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15525344

ABSTRACT

Recently, erythropoietin (EPO) and the nonerythropoietic derivative asialoEPO have been linked to tissue protection in the nervous system. In this study, we tested their effects in a model of neonatal hypoxia-ischemia (HI) in 7-day-old rats (unilateral carotid ligation and exposure to 7.7% O(2) for 50 min). EPO (10 U/g body weight = 80 ng/g; n = 24), asialoEPO (80 ng/g; n = 23) or vehicle (phosphate-buffered saline with 0.1% human serum albumin; n = 24) was injected intraperitoneally 4 h before HI. Both drugs were protective, as judged by measuring the infarct volumes, neuropathological score and gross morphological score. The infarct volumes were significantly reduced by both EPO (52%) and asialoEPO (55%) treatment, even though the plasma levels of asialoEPO had dropped below the detection limit (1 pm) at the onset of HI, while those of EPO were in the nanomolar range. Thus, a brief trigger by asialoEPO before the insult appears to be sufficient for protection. Proteomics analysis after asialoEPO treatment alone (no HI) revealed at least one differentially up-regulated protein, synaptosome-associated protein of 25 kDa (SNAP-25). Activation (phosphorylation) of ERK was significantly reduced in asialoEPO-treated animals after HI. EPO and the nonerythropoietic asialoEPO both provided significant and equal neuroprotection when administered 4 h prior to HI in 7-day-old rats. The protection might be related to reduced ERK activation and up-regulation of SNAP-25.


Subject(s)
Asialoglycoproteins/therapeutic use , Erythropoietin/analogs & derivatives , Erythropoietin/therapeutic use , Hypoxia-Ischemia, Brain/drug therapy , Animals , Animals, Newborn , Asialoglycoproteins/biosynthesis , Asialoglycoproteins/blood , Asialoglycoproteins/pharmacokinetics , Brain Infarction/pathology , Brain Infarction/prevention & control , Disease Models, Animal , Erythropoietin/biosynthesis , Erythropoietin/blood , Erythropoietin/pharmacokinetics , Female , Hypoxia-Ischemia, Brain/pathology , Male , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Proteomics/methods , Rats , Rats, Wistar , Signal Transduction/drug effects , Synaptosomal-Associated Protein 25 , Treatment Outcome
10.
J Drug Target ; 9(2): 155-68, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11697109

ABSTRACT

In this study, a small triantennary asialoglycopeptide of fetuin (A-F2) was used as a ligand to direct liposomes to hepatocytes. A-F2 was cleaved from asialofetuin, purified, conjugated with fatty acids and incorporated into pre-formed sonicated DSPC/Chol (2:1) liposomes. A mild cholate incubation method for incorporating the A-F2 ligand on pre-formed vesicles was used. In preliminary in vivo experiments 111In3+ encapsulated in A-F2/palmityl liposomes was seen to accumulate in the liver of mice significantly faster than when encapsulated in non-ligand bearing liposomes of the same lipid composition (studied before), justifying further investigation of this system. The presence of the A-F2/fatty acid conjugate in a functional form on the vesicle surface was confirmed by their reversible agglutination in the presence of Ricinus communis agglutinin (RCA120). Effects of ligand incorporation on the vesicle size distribution, z-potential, membrane integrity and stability were monitored. The results demonstrate that highest ligand incorporation was achieved when liposomes and ligand were co-incubated in the presence of 1 mM sodium cholate. Incorporation increased with the length of the fatty acid used for A-F2 conjugation. Ligand-bearing liposomes were demonstrated to be smaller in diameter (about 30%) with a more positive z-potential in comparison to control vesicles while ligand incorporation did not influence the liposome membrane integrity. The size of the ligand-incorporating vesicles was maintained after 24 hours of incubation in isotonic buffer, proving that the vesicles do not aggregate. Although the preliminary biodistribution results may suggest that ligand bearing liposomes are accumulating in the liver, further cell culture, in vivo distribution and especially liver fractionation studies are required in order to clarify the intrahepatic localization of these liposomes and the ability to target liver hepatocytes in vivo.


Subject(s)
Asialoglycoproteins/pharmacokinetics , alpha-Fetoproteins/pharmacokinetics , Animals , Asialoglycoproteins/chemistry , Cholesterol , Cholic Acids , Drug Carriers , Fatty Acids/chemistry , Fetuins , Fluorescent Dyes , Glycopeptides/chemistry , Glycopeptides/pharmacokinetics , Hepatocytes/drug effects , Ligands , Liposomes , Mice , Particle Size , Phosphatidylcholines , Tissue Distribution , alpha-Fetoproteins/chemistry
11.
Atherosclerosis ; 153(1): 219-30, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11058718

ABSTRACT

Desialylated low density lipoprotein (LDL) is rapidly taken up and accumulated by both peripheral blood monocytes and cells isolated from human arterial intima consisting predominantly of smooth muscle cells. It is shown that thioglycollate (TG)-elicited mouse macrophages and mouse peritoneal macrophages stimulated with lipopolysaccharide (LPS) show increased expression of a membrane-bound, galactose-specific lectin that could be responsible for this uptake. In LPS-stimulated macrophages accumulation of desialylated LDL is increased ca. 2.6-fold. Accumulation of acetylated LDL in the same cells is reduced, suggesting that the galactose-specific lectin might be responsible for the uptake of desialylated LDL. Transfection of cells with the mouse macrophage Gal/GalNAc-specific lectin (MMGL) increased their capacity to take up asialofetuin (ASF) and, to a smaller extent, desialylated LDL. The uptake of desialylated LDL was small, most likely due to the high k(d) of MMGL for biantennary oligosaccharides as found on LDL, and low concentration of LDL achieved in tissue culture experiments. The data suggest that the expression of galactose-specific lectins can be elevated under inflammatory conditions, and that these receptors could contribute to foam cell formation under conditions of high desialylated LDL concentration, as might be found in arterial intima.


Subject(s)
Galactose/metabolism , Lectins, C-Type , Lectins/physiology , Lipoproteins, LDL/pharmacokinetics , Macrophages/metabolism , Membrane Proteins , Animals , Asialoglycoproteins/pharmacokinetics , Carrier Proteins/pharmacology , Cell Line , Fetuins , Fluorescent Antibody Technique, Indirect , Humans , Lectins/metabolism , Lectins/pharmacology , Lipopolysaccharides/pharmacology , Lipoproteins, LDL/metabolism , Mice , N-Acetylneuraminic Acid/metabolism , Thioglycolates/pharmacology , Transfection , alpha-Fetoproteins/pharmacokinetics
12.
Eur J Biochem ; 267(21): 6501-8, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11029595

ABSTRACT

In mammals, clearance of desialylated serum glycoproteins to the liver is mediated by a galactose-specific hepatic lectin, the 'asialoglycoprotein receptor'. In humans, serum glycoprotein glycans are usually capped with sialic acid, which protects these proteins against hepatic uptake. However, in most other species, an additional noncharged terminal element with the structure Galalpha1-->3Galbeta1-->4R is present on glycoprotein glycans. To investigate if alpha3-galactosylated glycoproteins, just like desialylated glycoproteins, could be cleared by the hepatic lectin, the affinities of alpha3-galactosylated compounds towards this lectin were determined using an in vitro inhibition assay, and were compared with those of the parent compounds terminating in Galbeta1-->4R. Diantennary, triantennary and tetraantennary oligosaccharides that form part of N-glycans were alpha3-galactosylated to completion by use of recombinant bovine alpha3-galactosyltransferase. Similarly, desialylated alpha1-acid glycoprotein (orosomucoid) was alpha3-galactosylated in vitro. The alpha3-galactosylation of a branched, Galbeta1-->4-terminated oligosaccharide lowered its affinity for the membrane-bound lectin on whole rat hepatocytes 50-250-fold, and for the detergent-solubilized hepatic lectin 7-50-fold. In contrast, alpha3-galactosylation of asialo-alpha1-acid glycoprotein caused only a minor decrease in affinity, increasing the IC50 from 5 to 15 nM. Fully alpha3-galactosylated alpha1-acid glycoprotein, intravenously injected into the mouse, was rapidly cleared from the circulation, with a clearance rate close to that of asialo-alpha1-acid glycoprotein (t1/2 of 0.42 min vs. 0.95 min). Its uptake was efficiently inhibited by pre-injection of an excess asialo-fetuin. Organ distribution analysis showed that the injected alpha1-acid glycoprotein accumulated predominantly in the liver. Taken together, these observations suggest that serum glycoproteins that are heavily alpha3-galactosylated will be rapidly cleared from the bloodstream via the hepatic lectin. It is suggested that glycosyltransferase expression in murine hepatocytes is tightly regulated in order to prevent undesired uptake of hepatocyte-derived, circulating glycoproteins.


Subject(s)
Galactosyltransferases/metabolism , Glycoproteins/chemistry , Glycoproteins/metabolism , Liver/chemistry , Orosomucoid/analogs & derivatives , Receptors, Cell Surface/metabolism , Animals , Asialoglycoprotein Receptor , Asialoglycoproteins/isolation & purification , Asialoglycoproteins/metabolism , Asialoglycoproteins/pharmacokinetics , Blood Proteins/metabolism , Blood Proteins/pharmacokinetics , Cattle , Galactose/metabolism , Glycoproteins/isolation & purification , Glycosylation , Half-Life , Hepatocytes/chemistry , Hepatocytes/metabolism , Humans , Inhibitory Concentration 50 , Lectins/metabolism , Liver/cytology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Oligosaccharides/chemistry , Oligosaccharides/isolation & purification , Oligosaccharides/metabolism , Orosomucoid/isolation & purification , Orosomucoid/metabolism , Orosomucoid/pharmacokinetics , Protein Binding , Rats
13.
Am J Physiol ; 277(6): G1189-99, 1999 12.
Article in English | MEDLINE | ID: mdl-10600816

ABSTRACT

Fibronectin (Fn) is a major adhesive protein found in the hepatic extracellular matrix (ECM). In adult rats, the in vivo turnover of plasma Fn (pFn) incorporated into the liver ECM is relatively rapid, i.e., <24 h, but the regulation of its turnover has not been defined. We previously reported that cellular Fn (cFn) and enzymatically desialylated plasma Fn (aFn), both of which have a high density of exposed terminal galactose residues, rapidly interact with hepatic asialoglycoprotein receptors (ASGP-R) in association with their plasma clearance after intravenous infusion. With the use of adult male rats (250-350 g) and measurement of the deoxycholate (DOC)-insoluble (125)I-labeled Fn in the liver, we determined whether the ASGP-R system can also influence the hepatic matrix retention of various forms of Fn. There was a rapid deposition of (125)I-pFn, (125)I-aFn, and (125)I-cFn into the liver ECM after their intravenous injection. Although (125)I-pFn was slowly lost from the liver matrix over 24 h, more than 90% of the incorporated (125)I-aFn and (125)I-cFn was cleared within 4 h (P < 0.01). Intravenous infusion of excess nonlabeled asialofetuin to competitively inhibit the hepatic ASGP-R delayed the rapid turnover of both aFn and cFn already incorporated within the ECM of the liver. ECM retention of both (125)I-aFn and (125)I-cFn was also less than (125)I-pFn (P < 0.01) as determined in vitro using liver slices preloaded in vivo with either tracer form of Fn. The hepatic ASGP-R appears to participate in the turnover of aFn and cFn within the liver ECM, whereas a non-ASGP-R-associated endocytic pathway apparently influences the removal of normal pFn incorporated within the hepatic ECM, unless it becomes locally desialylated.


Subject(s)
Extracellular Matrix Proteins/metabolism , Fibronectins/pharmacokinetics , Liver/metabolism , Receptors, Cell Surface/metabolism , Animals , Antirheumatic Agents/pharmacology , Asialoglycoprotein Receptor , Asialoglycoproteins/isolation & purification , Asialoglycoproteins/metabolism , Asialoglycoproteins/pharmacokinetics , Cell Fractionation , Chloroquine/pharmacology , Cross-Linking Reagents/metabolism , Cross-Linking Reagents/pharmacokinetics , Deoxycholic Acid , Detergents , Endocytosis/physiology , Fetuins , Fibroblasts/chemistry , Fibronectins/chemistry , Fibronectins/metabolism , Galactose/metabolism , Humans , Iodine Radioisotopes , Isomerism , Liver/chemistry , Liver/drug effects , Male , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , alpha-Fetoproteins/isolation & purification , alpha-Fetoproteins/metabolism , alpha-Fetoproteins/pharmacokinetics
14.
Z Gastroenterol ; 34 Suppl 3: 79-80, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8767471

ABSTRACT

The aim of this study was to demonstrate the influence of endocytosed asialoorosomucoid gold complexes (ASOR-Au10, 10 = diameter of gold particles) on the density of endocytic compartments in rat hepatocytes when fractionated on linear sucrose gradients. We show that by loading hepatocytes with ASOR-Au10 for different time intervals, organelles containing gold particles are denser than organelles containing unconjugated asialoorosomucoid (ASOR). This density shift was observed in endosomes, which were loaded with a short pulse (15 minutes) of ASOR- Au10. Also lysosomes, loaded with ASOR-Au10 for 120 minutes at 37 degrees C, display a similar density shift.


Subject(s)
Centrifugation, Density Gradient , Coated Vesicles/physiology , Endocytosis/physiology , Gold Colloid/pharmacokinetics , Animals , Asialoglycoproteins/pharmacokinetics , Cells, Cultured , Liver/cytology , Organelles/metabolism , Orosomucoid/analogs & derivatives , Orosomucoid/pharmacokinetics , Rats
15.
Magn Reson Imaging ; 13(7): 991-1002, 1995.
Article in English | MEDLINE | ID: mdl-8583878

ABSTRACT

Testing of receptor-specific MR contrast agents targeted to the liver is hampered by a shortage of viable in vitro models with in vivo-like hepatocellular morphology. Coated pits are ultrastructural signs of an active receptor mediated endocytosis in hepatocytes. Expression of coated pits by matrix overlaid hepatocytes was studied by transmission electron microscopy. Binding of a rhodaminated asialoglycoprotein receptor agent (MION-ASF-rh) was assessed by fluorescence microscopy. Fluorescence of cells exposed to MION-ASF-rh with D(+)-galactose reduced fluorescent light emission to a level of 58% of MION-ASF-rh-induced fluorescence. After preincubation with the hepatotoxin CCl4 a dose-dependent decrease in fluorescent light emission resulted. Hepatocytes maintained a homogeneous cell surface expression, with microprojections, coated pits, and vesicles on both sinusoidal surfaces. Matrix overlaid primary hepatocytes constitute a viable, morphologically and functionally differentiated model. This model can be used to study receptor binding, uptake, and blockage of diagnostic magnetopharmaceuticals under controlled conditions.


Subject(s)
Asialoglycoproteins/metabolism , Contrast Media , Ferric Compounds , Iron , Liver/cytology , Magnetic Resonance Imaging , Oxides , Receptors, Cell Surface/metabolism , Rhodamines , alpha-Fetoproteins , Animals , Asialoglycoprotein Receptor , Asialoglycoproteins/pharmacokinetics , Cells, Cultured , Female , Ferric Compounds/pharmacokinetics , Ferrosoferric Oxide , Fetuins , Iron/pharmacokinetics , Liver/metabolism , Liver/ultrastructure , Microscopy, Electron , Oxides/pharmacokinetics , Rats , Rats, Wistar , Rhodamines/pharmacokinetics , alpha-Fetoproteins/pharmacokinetics
16.
Liver ; 14(3): 141-7, 1994 Jun.
Article in English | MEDLINE | ID: mdl-7521505

ABSTRACT

Liver sinusoidal cells play an important role in host defense by clearing particulate matter and macromolecules from the circulation. In this study, receptor-mediated endocytosis in sinusoidal cells was examined in two-thirds hepatectomized rats using 125I-labeled formaldehyde-treated bovine serum albumin (fBSA) as an endocytable macromolecule. The liver-weight to body-weight ratio in hepatectomized rats returned to the control value 10 days after hepatectomy. The endocytotic index for fBSA in sinusoidal cells decreased significantly to 0.0210 +/- 0.0017 (controls, 0.0598 +/- 0.0019) on the first day, then returned to the control level at 5 days (0.0554 +/- 0.0030). The changes in hepatic uptake for fBSA showed a similar time course of the endocytotic index. A transient increase in the uptake of fBSA per unit weight of liver of 22-39% above control occurred 2 to 3 days after hepatectomy. In contrast to fBSA, the endocytotic index in hepatocytes evaluated with 125I-labeled asialofetuin reached the minimum level on the second day, and then recovered to the control level 10 days after hepatectomy. These results suggest that endocytosis of fBSA by sinusoidal cells decreases after hepatectomy and rapidly recovers to normal before the completion of liver regeneration, whereas endocytosis of asialofetuin by hepatocytes decreases following hepatic resection and returns to normal when regeneration is substantially complete.


Subject(s)
Endocytosis/physiology , Endothelium, Vascular/cytology , Kupffer Cells/physiology , Liver Regeneration/physiology , Liver/cytology , Animals , Asialoglycoproteins/pharmacokinetics , Endothelium, Vascular/physiology , Fetuins , Formaldehyde/pharmacokinetics , Hepatectomy , Iodine Radioisotopes , Liver/physiology , Male , Rats , Rats, Wistar , Serum Albumin, Bovine/pharmacokinetics , Time Factors , alpha-Fetoproteins/pharmacokinetics
17.
Lab Invest ; 69(6): 714-23, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8264233

ABSTRACT

BACKGROUND: Whereas the complex removal routes hypothesized for IgA containing immune complexes (IC) and macromolecules can be adequately analyzed by a recently proposed IgA1-IgG aggregate probe (Lab Invest, 66: 86-95), the relative significance of the asialoglycoprotein receptors in IgAIC clearance is still uncertain. EXPERIMENTAL DESIGN: The removal kinetics of 99mTc diethylenetriamine-pentaacetic acid-conjugated asialo alpha 1 acid glycoprotein (AAGP) and 123I-labeled IgA1-IgG aggregate were analyzed in 11 cirrhosis patients and 13 IgAN patients of comparable age. RESULTS: IgA1-IgG aggregate mean plasma clearance rate was delayed in IgA neuropathy (IgAN) patients (slope 0.038 minutes-1, range 0.027 to 0.053) compared with normals (0.047 minutes-1, range 0.038 to 0.053, p = 0.05). The liver was the main organ involved in the IgA1-IgG removal. When compared with normals, (34.3 minutes, range 29.8 to 42.2), the liver mean transit time (MTT) was significantly (p < 0.02) prolonged in IgAN patients (41.3 minutes, 33.6 to 52.3). Participation of spleen in clearance was observed in some patients and was almost invariably concurrent with normal clearance parameters. Conversely, 9 out of 11 cirrhosis patients had a remarkable splenic uptake, but the blood clearance rate was invariably delayed (0.022 minutes-1, 0.014 to 0.028, p < 0.003) and liver MTT extremely prolonged (122.4 minutes, 52.4 to 400, p < 0.003). In IgAN patients with delayed clearance of the IgA1-IgG aggregate, a distinct trend of progression towards renal failure was noted. AAGP clearance was also delayed in cirrhosis patients: slope = 0.166 minutes-1, 0.108 to 0.247, p = 0.05 as compared with both normals (0.230, 0.173 to 0.289) and IgAN patients (0.250, 0.184 to 0.254). Liver MTT in cirrhosis patients was extremely prolonged: 240.6 minutes, 132.5 to 400 minutes, p < 0.007 compared with both normals (90.0 minutes, 82.7 to 96.6) and IgA patients (92.2 minutes, 70.3 to 107.1). AAGP clearance parameters in normals and IgAN patients were not statistically different. MTT values of AAGP and IgA1-IgG aggregate were strictly related (p = 0.008), suggesting that asialoglycoprotein receptors are partially involved in the clearance of the IgA1-IgG aggregate probe. CONCLUSIONS: Some patients with IgAN have a prolonged circulation of an IgAIC miming probe, probably due to an impaired macrophage function. Other possibilities of prolonged circulation of IgAIC in these patients should imply an abnormal IgA glycosylation pattern that allows IC to escape from an effective asialoglycoprotein receptor system. In cirrhosis patients, all of the removal routes of IgA and IgA containing IC are greatly altered suggesting a causative role in the development of an associated, often clinically inapparent, glomerular disease.


Subject(s)
Antigen-Antibody Complex/metabolism , Glomerulonephritis, IGA/metabolism , Immunoglobulin A/metabolism , Liver Cirrhosis, Alcoholic/metabolism , Receptors, Cell Surface/metabolism , Adult , Aged , Asialoglycoprotein Receptor , Asialoglycoproteins/metabolism , Asialoglycoproteins/pharmacokinetics , Female , Humans , Male , Metabolic Clearance Rate , Middle Aged , Orosomucoid/analogs & derivatives , Orosomucoid/metabolism , Orosomucoid/pharmacokinetics , Radionuclide Imaging
18.
Magn Reson Imaging ; 11(3): 411-7, 1993.
Article in English | MEDLINE | ID: mdl-7685055

ABSTRACT

Receptor-directed MR contrast agents are currently being designed to improve sensitivity and specificity of MR imaging and to provide for functional MR imaging. In the current study we have synthesized a conjugate of asialofetuin (ASF), a bovine plasma protein with a known, high affinity for the hepatic asialoglycoprotein receptor, and a well defined, single crystal superparamagnetic label (monocrystalline iron oxide nanoparticle, MION). MION-ASF is cleared from the circulation more than 300 times faster than MION, has a 3.7 times higher hepatic accumulation, increases liver R2 relaxivity 2.8-fold compared to MION, and accumulates in hepatocytes unlike MION, which accumulates only in macrophages. Competition assays indicate that receptor-mediated hepatocyte uptake can be competitively blocked and that this effect can be demonstrated by imaging. These studies indicate that sensitive iron oxide based probes can be developed for functional MR imaging.


Subject(s)
Asialoglycoproteins/pharmacokinetics , Contrast Media/pharmacokinetics , Ferric Compounds/pharmacokinetics , Magnetic Resonance Imaging , alpha-Fetoproteins/pharmacokinetics , Animals , Asialoglycoprotein Receptor , Asialoglycoproteins/metabolism , Autoradiography , Fetuins , Indium Radioisotopes , Liver/diagnostic imaging , Liver/metabolism , Male , Radionuclide Imaging , Rats , Rats, Sprague-Dawley , Receptors, Immunologic/metabolism
19.
Biochim Biophys Acta ; 942(1): 57-64, 1988 Jul 07.
Article in English | MEDLINE | ID: mdl-3382658

ABSTRACT

Isolated hepatocytes of rat, rabbit and guinea pig were found to take up and degrade 125I-labelled asialoorosomucoid at different rates with the rank order: rabbit greater than rat greater than guinea pig. Measurement of 125I-asialoorosomucoid binding at 4 degrees C to these hepatocytes revealed that all these cells had two classes of receptors with a major difference occurring in the number of high-affinity binding sites. The average binding affinity constants (K) and receptor concentration (N) calculated from a least-square analysis of the Scatchard plots were K1 = 1.15.10(9) M-1, K2 = 0.93.10(7) M-1, N1 = 0.049 pmol/mg cell protein and N2 = 0.27 pmol/mg cell protein for the rat; K2 = 3.16.10(7) M-1, N1 = 0.027 pmol/mg cell protein and N2 = 0.13 pmol/mg cell protein for the guinea pig and K1 = 0.74.10(9) M-1, K2 = 3.85.10(7) M-1, N1 = 0.205 pmol/mg cell protein and N2 = 0.37 pmol/mg cell protein for the rabbit hepatocytes, respectively. Measurement of the total number of cellular receptors after solubilization with Triton X-100 also revealed the same receptor concentration rank order of rabbit (5.8 pmol/mg cell protein) greater than rat (0.55 pmol/mg cell protein) greater than guinea pig (0.18 pmol/mg cell protein). Intravenous injection of 125I-asialoorosomucoid into anesthetized animals of matched body weight also indicated that the rate of plasma clearance and the rate of appearance of the degraded product of the tracer were different among these species with the same rank order as that observed with isolated hepatocytes. Thus there is a fundamental difference in the number of asialoglycoprotein receptors both on the cell surface and inside hepatocytes of these mammalian species.


Subject(s)
Asialoglycoproteins/metabolism , Liver/metabolism , Receptors, Immunologic/metabolism , Animals , Asialoglycoprotein Receptor , Asialoglycoproteins/pharmacokinetics , Biological Transport , Guinea Pigs , Metabolic Clearance Rate , Rabbits , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...