Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Pharm Sci ; 101(4): 1599-606, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22081433

ABSTRACT

Human α(1) -acid glycoprotein (AGP), a serum glycoprotein, is known to have anti-inflammatory activity. We recently reported that AGP was mainly incorporated into the liver in mice via a receptor-mediated pathway, although the mechanism for this was largely unknown. The objective of this study was to identify the specific cellular surface protein that recognizes the peptide moiety of AGP. Pharmacokinetic studies of (111) In-AGP and (111) In -recombinant glycan-deficient AGP (rAGP) in mice demonstrated that both AGPs are mainly distributed to the liver and kidney, but hepatic and renal uptake clearance of rAGP was higher than that for AGP. Hepatic uptake of rAGP was inhibited in the presence of 100-fold excess of unlabeled AGP, indicating that the hepatic uptake of rAGP shared a common route with that of AGP and that it recognized the peptide moiety of AGPs. In ligand blotting analyses using crude cellular membrane fraction of mice liver, a band corresponding to a 16 kDa protein was observed to bind to both AGPs. Interestingly, matrix-assisted laser desorption ionization-time-of-flight mass spectrometry MALDI-TOF-MS and western blotting analyses indicated that this 16 kDa protein is the hemoglobin ß-chain (HBB). It, therefore, appears that HBB is associated with the hepatic uptake of AGP via a direct interaction with its peptide moiety.


Subject(s)
Hemoglobins/metabolism , Liver/metabolism , Orosomucoid/pharmacokinetics , Animals , Hep G2 Cells , Hepatocytes/metabolism , Humans , Male , Mice , Orosomucoid/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
2.
Free Radic Res ; 45(10): 1196-206, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21781009

ABSTRACT

Endogenous S-nitrosated human serum albumin (E-Mono-SNO-HSA) is a large molecular weight nitric oxide (NO) carrier in human plasma, which has shown many beneficial effects in different animal models. To construct more efficient SNO-HSA preparations, SNO-HSA with many conjugated SNO groups has been prepared using chemical modification (CM-Poly-SNO-HSA). We have compared the properties of such a preparation to those of E-Mono-SNO-HSA. Cellular uptake of NO from E-Mono-SNO-HSA partly takes place via low molecular weight thiol, and it results in cytoprotective effects by induction of heme oxygenase-1. By contrast, transfer of NO from CM-Poly-SNO-HSA into the cells is faster and more pronounced. The influx mainly takes place by cell-surface protein disulfide isomerase. The considerable NO inflow results in apoptotic cell death by ROS induction and caspase-3 activation. Thus, increasing the number of SNO groups on HSA does not simply intensify the cellular responses to the product but can also result in very different effects.


Subject(s)
Nitric Oxide/metabolism , Nitroso Compounds/chemical synthesis , Nitroso Compounds/metabolism , Serum Albumin/chemical synthesis , Serum Albumin/metabolism , Animals , Cell Line, Tumor , Cysteine/chemistry , Cysteine/metabolism , Hep G2 Cells , Humans , Mice , Nitric Oxide/chemistry , Nitrosation , Protein Disulfide-Isomerases/metabolism , Serum Albumin/chemistry , Serum Albumin, Human
3.
J Control Release ; 147(1): 17-23, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20678999

ABSTRACT

Thioredoxin-1 (Trx) is a redox-active protein with anti-inflammatory effects. The effect of albumin fusion on the pharmacokinetic and pharmacodynamic properties of Trx was evaluated in this study. The findings indicate that the properties of human serum albumin and the fusion protein are comparable. The fusion protein showed similar plasma concentration and organ distribution profiles as human serum albumin. The fusion protein accumulated in lungs, reaching levels higher than Trx. In an insulin reducing assay, the activity of the fusion protein was 60% of the activity of Trx. However, survival rate of endotoxic shock mice induced by the administration of a lipopolysaccharide and D-galactosamine for fusion protein was double that of Trx. The findings reported herein indicate that the fusion protein is likely to have great clinical applications in areas such as the treatment of reperfusion injuries.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal , Recombinant Fusion Proteins , Serum Albumin/chemistry , Thioredoxins , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Blotting, Western , Circular Dichroism , Disease Models, Animal , Drug Stability , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Humans , Male , Mice , Mice, Inbred Strains , Pichia/genetics , Protein Conformation , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/therapeutic use , Serum Albumin/genetics , Shock, Septic/drug therapy , Survival Analysis , Thioredoxins/administration & dosage , Thioredoxins/pharmacokinetics , Thioredoxins/therapeutic use , Tissue Distribution
4.
J Control Release ; 145(1): 9-16, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20304018

ABSTRACT

Human serum albumin (HSA), a non-glycosylated protein, is widely employed as carrier for drug delivery systems. A series of recombinant, mannosylated-HSA mutants (Man-rHSAs: D63N, A320T and D494N) and their triple mutant (TM-rHSA: D63N/A320T/D494N) were prepared, that can be selectively delivered to the liver via mannose receptor (MR) on the liver non-parenchymal cells. A pharmacokinetic analysis of (111)In-Man-rHSAs in mice showed that they were rapidly cleared from the blood circulation, and were largely taken up by the liver rapidly in the order: TM-rHSA>D494N>>A320T=D63N, consistent with their degree of mannosylation. In vivo competition experiments with an excess amount of chemically modified Man-BSA or mannan suggested that the hepatic uptake of TM-rHSA was selectively mediated by MR on Kupffer cells. Lastly, a TM-rHSA-NO conjugate, S-nitrosylated TM-rHSA, effectively delivered NO to the liver and then exhibited a significant inhibitory effect against hepatic ischemia/reperfusion injury model rats, accompanied by the induction of heme oxygenase-1.


Subject(s)
Drug Carriers/chemistry , Lectins, C-Type/metabolism , Liver/drug effects , Mannose-Binding Lectins/metabolism , Mannose/genetics , Receptors, Cell Surface/metabolism , Recombinant Proteins/genetics , Serum Albumin/genetics , Animals , Blotting, Western , Circular Dichroism , Disease Models, Animal , Drug Carriers/pharmacokinetics , Electrophoresis, Polyacrylamide Gel , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Heme Oxygenase-1/metabolism , Humans , Kupffer Cells/drug effects , Kupffer Cells/metabolism , Liver/blood supply , Liver/metabolism , Mannose/chemistry , Mannose/pharmacokinetics , Mannose Receptor , Mice , Mice, Inbred Strains , Nitric Oxide/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacokinetics , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Serum Albumin/chemistry , Serum Albumin/pharmacokinetics , Tissue Distribution
5.
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.
Nitric Oxide ; 22(4): 259-65, 2010 May 15.
Article in English | MEDLINE | ID: mdl-19818866

ABSTRACT

Recently, nitric oxide has been investigated as a potential anti-cancer therapy because of its cytotoxic activity. Previously, we found that S-nitrosylated human serum albumin (SNO-HSA) induced apoptosis in C26 cells, demonstrating for the first time that SNO-HSA has potential as an anti-cancer drug. In the present study, the anti-tumor activity of SNO-HSA in another tumor type of cancer cell was investigated using murine tumor LY-80 cells. Mitochondrial depolarization, activation of caspase-3 and DNA fragmentation were induced in LY-80 cells by SNO-HSA treatment in a dose-dependent manner. Inhibition of caspase activity resulted in complete inhibition of DNA fragmentation induced by SNO-HSA. The cytotoxic effects of SNO-HSA on LY-80 were concentration-dependent. Tumor growth in LY-80-tumor-bearing rats was significantly inhibited by administration of SNO-HSA compared with saline- and HSA-treatment. These results suggest that SNO-HSA has potential as a chemopreventive and/or chemotherapeutic agent because it induces apoptosis in tumor cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Nitroso Compounds/pharmacology , Serum Albumin/pharmacology , Animals , Caspase 3/metabolism , Cell Line, Tumor , Cell Survival , Humans , Male , Nitric Oxide/toxicity , Rats , Rats, Inbred Strains , Serum Albumin, Human
7.
Am J Physiol Renal Physiol ; 295(6): F1871-80, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18971214

ABSTRACT

Chronic accumulation of plasma advanced oxidation protein products (AOPPs) promotes renal fibrosis. However, the mechanism at the cellular level has not been clarified. In the present study, endocytic assay of human proximal tubular cells (HK-2 cells) demonstrated that AOPPs-human serum albumin (HSA) (in vitro preparations of chloramine- modified HSA) were significantly endocytosed in a dose-dependent manner at a higher level than HSA. The expression of CD36, a transmembrane protein of the class B scavenger receptor, in HK-2 cells was confirmed in the immunoblot analysis. In a cellular assay using overexpressing human CD36 in Chinese hamster ovary (CHO) cells, AOPPs-HSA were significantly endocytosed by CD36-CHO cells but not by mock-CHO cells. Furthermore, the endocytic association and degradation of AOPPs-HSA by HK-2 cells was significantly inhibited by anti-CD36 antibody treatment, suggesting that CD36 is partly involved in the uptake of AOPPs-HSA by HK-2 cells. AOPPs-HSA upregulated the expression of CD36 in a dose-dependent manner. In addition, AOPPs-HSA upregulated the generation of intracellular reactive oxygen species and the secretion of transforming growth factor (TGF)-beta1 in HK-2 cells, whereas anti-CD36 antibody neutralizes the upregulation of TGF-beta1. These results suggest that AOPPs-HSA may cause renal tubular injury via the CD36 pathway.


Subject(s)
CD36 Antigens/physiology , Kidney Tubules/injuries , Proteins/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Endocytosis/physiology , Fibrosis , Humans , Kidney/pathology , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Serum Albumin/pharmacology
8.
J Pharmacol Exp Ther ; 325(1): 69-76, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18218831

ABSTRACT

In recent studies, the cytotoxic activity of NO has been investigated for its potential use in anticancer therapies. Nitrosated human serum albumin (NO-HSA) may act as a reservoir of NO in vivo. However, there are no published reports regarding the effects of NO-HSA on cancer. Therefore, the present study investigated the antitumor activity of NO-HSA. NO-HSA was prepared by incubating HSA, which had been sulfhydrylated using iminothiolane, with isopentyl nitrite (6.64 mol NO/mol HSA). Antitumor activity was examined in vitro using murine colon 26 carcinoma (C26) cells and in vivo using C26 tumor-bearing mice. Exposure to NO-HSA increased the production of reactive oxygen species in C26 cells. Flow cytometric analysis using rhodamine 123 showed that NO-HSA caused mitochondrial depolarization. Activation of caspase-3 and DNA fragmentation were observed in C26 cells after incubation with 100 muM NO-HSA for 24 h, and NO-HSA inhibited the growth of C26 cells in a concentration-dependent manner. The growth of C26 tumors in mice was significantly inhibited by administration of NO-HSA compared with saline and HSA treatment. Immunohistochemical analysis of tumor tissues demonstrated an increase in terminal deoxynucleotidyl transferase dUTP nickend labeling-positive cells in NO-HSA-treated mice, suggesting that inhibition of tumor growth by NO-HSA was mediated through induction of apoptosis. Biochemical parameters (such as serum creatinine, blood urea nitrogen, aspartate aminotransferase, and alanine aminotransferase) showed no significant differences among the three treatment groups, indicating that NO-HSA did not cause hepatic or renal damage. These results suggest that NO-HSA has the potential for chemopreventive and/or chemotherapeutic activity with few side effects.


Subject(s)
Neoplasms, Experimental/drug therapy , Nitroso Compounds/therapeutic use , Serum Albumin/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Design , Humans , Immunohistochemistry , Mice , Neoplasms, Experimental/pathology , Nitroso Compounds/chemical synthesis , Nitroso Compounds/pharmacology , Serum Albumin/chemical synthesis , Serum Albumin/pharmacology , Serum Albumin, Human , Treatment Outcome
9.
Biochim Biophys Acta ; 1772(11-12): 1192-8, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18053692

ABSTRACT

The cellular interaction of proteins modified with advanced glycation end-products (AGEs) is believed to induce several different biological responses, which are involved in the development of diabetic vascular complications. We report here that the ratio of protein glycation is implicated in its ligand activity to scavenger receptors. Although highly-modified AGE-bovine serum albumin (high-AGE-BSA) was significantly recognized by human monocyte-derived macrophages and Chinese hamster ovary cells which overexpress such scavenger receptors as CD36, SR-BI (scavenger receptor class B type-I), and LOX-1 (Lectin-like Ox-LDL receptor-1), the mildly-modified-AGE-BSA (mild-AGE-BSA) did not show any ligand activity to these cells. Furthermore, when (111)In-labeled high- or mild-AGE-BSA were injected into the tail vein of mice, the high-AGE-BSA was rapidly cleared from the circulation whereas the clearance rate of the mild-AGE-BSA was very slow, similar to the native BSA. These results demonstrate the first evidence that the ligand activity of the AGE-proteins to the scavenger receptors and its pharmacokinetic properties depend on their rate of modification by AGEs, and we should carefully prepare the AGE-proteins in vitro to clarify the physiological significance of the interaction between the AGE-receptors and AGE-proteins.


Subject(s)
Glycation End Products, Advanced/metabolism , Receptors, Scavenger/metabolism , Aldehydes/metabolism , Animals , CHO Cells , Cattle , Chromatography, High Pressure Liquid , Cricetinae , Cricetulus , Crystallins/metabolism , Glycation End Products, Advanced/administration & dosage , Glycation End Products, Advanced/chemistry , Glycation End Products, Advanced/pharmacology , Humans , Ligands , Lysine/analogs & derivatives , Lysine/metabolism , Macrophages/metabolism , Mice , Serum Albumin, Bovine/administration & dosage , Serum Albumin, Bovine/chemistry , Serum Albumin, Bovine/metabolism , Serum Albumin, Bovine/pharmacology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tissue Distribution
10.
J Mol Biol ; 363(3): 702-12, 2006 Oct 27.
Article in English | MEDLINE | ID: mdl-16979183

ABSTRACT

Human serum albumin is the most abundant protein in the circulatory system, and one of its principal functions is to transport fatty acids. Binding of octanoate, decanoate, laurate and myristate was studied by a rate-of-dialysis technique. The primary association constants increased, but not linearly, with chain length. The number of high-affinity sites also increased with chain length; octanoate and decanoate bind to one such site, whereas laurate and myristate most probably bind to two sites. Albumin is composed of three homologous helical domains (I-III), which can be subdivided into two subdomains (A and B). For getting information about the positions of the high-affinity sites we produced 13 recombinant isoforms mutated in four different subdomains. Results obtained with these albumins are in accordance with the following model: octanoate and decanoate bind to a single site in subdomain IIIA, laurate binds to sites in subdomains IIIA and IIIB, whereas myristate binds in subdomains IB and IIIB. The results also showed that primary fatty acid binding is sensitive to amino acid substitutions in other parts of the protein. This is in contrast to the effect of amino acid substitutions of genetic albumin variants (alloalbumins). Usually these substitutions, which are situated at the surface of the protein, have no effect on fatty acid binding. Binding of fatty acid anions to different high-affinity sites and the sensitivity of these sites to amino acid substitutions elsewhere in the protein (and perhaps also to other types of modifications) are important factors that could effect simultaneous binding of other ligands, e.g. in patients treated with albumin-binding drugs.


Subject(s)
Anions , Fatty Acids , Serum Albumin/chemistry , Serum Albumin/metabolism , Anions/chemistry , Anions/metabolism , Binding Sites , Fatty Acids/chemistry , Fatty Acids/metabolism , Humans , Models, Molecular , Mutagenesis, Site-Directed , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serum Albumin/genetics
11.
J Biochem ; 139(5): 821-9, 2006 May.
Article in English | MEDLINE | ID: mdl-16751589

ABSTRACT

Cellular interactions with advanced glycation end products (AGE)-modified proteins are known to induce several biological responses, not only endocytic uptake and degradation, but also the induction of cytokines and growth factors, combined responses that may be linked to the development of diabetic vascular complications. In this study we demonstrate that A549 cells, a human pulmonary epithelial cell line, possess a specific binding site for AGE-modified bovine serum albumin (AGE-BSA) (K(d) = 27.8 nM), and additionally for EN-RAGE (extracellular newly identified RAGE binding protein) (K(d) = 118 nM). Western blot and RT-PCR analysis showed that RAGE (receptor for AGE) is highly expressed on A549 cells, while the expression of other known AGE-receptors such as galectin-3 and SR-A (class A scavenger receptor), are below the level of detection. The binding of (125)I-AGE-BSA to these cells is inhibited by unlabeled AGE-BSA, but not by EN-RAGE. In contrast, the binding of (125)I-EN-RAGE is significantly inhibited by unlabeled EN-RAGE and soluble RAGE, but not by AGE-BSA. Our results indicate that A549 cells possess at least two binding sites, one specific for EN-RAGE and the other specific for AGE-BSA. The latter receptor on A549 cells is distinct from the scavenger receptor family and RAGE.


Subject(s)
Epithelial Cells/metabolism , Glycation End Products, Advanced/metabolism , Receptors, Immunologic/classification , Animals , Cattle , Cells, Cultured , Glycation End Products, Advanced/pharmacokinetics , Humans , Lung/metabolism , Protein Binding , Receptor for Advanced Glycation End Products , Receptors, Immunologic/agonists , Receptors, Scavenger/agonists , Receptors, Scavenger/classification , Receptors, Scavenger/metabolism , Recombinant Proteins/metabolism , Serum Albumin, Bovine/metabolism , Serum Albumin, Bovine/pharmacokinetics , Signal Transduction , Substrate Specificity
12.
Drug Metab Pharmacokinet ; 21(2): 140-6, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16702734

ABSTRACT

To determine the pharmacokinetic properties of advanced oxidation protein products (AOPP), we prepared oxidized human serum albumin (oxi-HSA) using chloramine-T (a hypochlorite analogue) in vitro. The AOPP and dityrosine content of oxi-HSA (AOPP content, 244.3+/-12.3 microM; dityrosine content, 0.7+/-0.11 nmol of dityrosine/mg protein) were similar to those of uremic patients. In structural analysis, the increases in AOPP and dityrosine content of HSA induced slight decreases in its alpha-helical content. In pharmacokinetic analysis, oxi-HSA left the circulation rapidly, and organ distribution of oxi-HSA 30 min after intravenous injection was 51% for the liver, 23% for the spleen, and 9% for the kidney, suggesting that the liver and spleen were the main routes of plasma clearance of oxi-HSA. The liver and spleen uptake clearance of oxi-HSA were significantly greater than those of normal HSA (CLliver, 5058+/-341.6 vs 24+/-4.2 microL/hr [p<0.01]; CLspleen, 2118+/-322.1 vs 32+/-2.7 microL/hr [p<0.01]). However, uptake by other organs was not significantly affected by oxidation. These results suggest that the liver and spleen play important roles in elimination of AOPP.


Subject(s)
Biological Products/pharmacokinetics , Serum Albumin/pharmacokinetics , Amino Acids/metabolism , Animals , Blotting, Western , Male , Mice , Oxidation-Reduction , Tyrosine/analogs & derivatives , Tyrosine/metabolism
13.
Biochem Biophys Res Commun ; 334(4): 1322-8, 2005 Sep 09.
Article in English | MEDLINE | ID: mdl-16054887

ABSTRACT

Oxidized albumin is a reliable marker of oxidative stress in hemodialysis (HD) patients. However, oxidized albumin in vivo and its possible clinical significance has been rarely investigated. In the present study, the qualitative modification of albumin in HD patients (n = 20) was examined and their results were compared with healthy age-matched controls (n = 10). The increase in plasma protein carbonyl levels in HD patients was largely due to an increase in oxidized albumin. Human serum albumin (HSA) of HD patients, HSA of HD patients (HD-HSA) and normal subjects (Normal-HSA) were purified on a blue Sepharose CL-6B column. Spectroscopic analysis confirmed that the HD-HSA samples contained higher levels of carbonyls than Normal-HSA. An HPLC analysis also suggested that the state of the purified HSA used throughout the experiments accurately reflects the redox state of albumin in blood. HD-HSA was found to have a decreased the antioxidant activity, and was able to trigger the oxidative burst of human neutrophils, compared to Normal-HSA. HD-HSA was conformationally altered, with its hydrophobic regions more exposed and to have a negative charge. In binding experiments, HD-HSA showed impaired Site II-ligand binding capabilities. Collectively, the oxidation of plasma proteins, especially HSA, might enhance oxidative stress in HD patients.


Subject(s)
Kidney Failure, Chronic/blood , Neutrophils/metabolism , Reactive Oxygen Species/blood , Renal Dialysis , Respiratory Burst , Serum Albumin/analysis , Serum Albumin/chemistry , Adult , Aged , Female , Humans , Kidney Failure, Chronic/rehabilitation , Male , Middle Aged , Oxidation-Reduction , Oxidative Stress , Protein Binding , Protein Conformation
14.
J Biochem ; 137(5): 607-16, 2005 May.
Article in English | MEDLINE | ID: mdl-15944414

ABSTRACT

Circulating proteins modified by advanced glycation end-products (AGE) are mainly taken up by liver endothelial cells (LECs) via scavenger receptor-mediated endocytosis. Endocytic uptake of chemically modified proteins by macrophages and macrophage-derived cells is mediated by class A scavenger receptor (SR-A) and CD36. In a previous study using SR-A knockout mice, we demonstrated that SR-A is not involved in endocytic uptake of AGE proteins by LECs [Matsumoto et al. (2000) Biochem. J. 352, 233-240]. The present study was conducted to determine the contribution of CD36 to this process. Glycolaldehyde-modified BSA (GA-BSA) and methylglyoxal-modified BSA (MG-BSA) were used as AGE proteins. 125I-GA-BSA and 125I-MG-BSA underwent endocytic degradation by these cells at 37 degrees C, and this process was inhibited by several ligands for the scavenger receptors. However, this endocytic uptake of 125I-GA-BSA by LECs was not inhibited by a neutralizing anti-CD36 antibody. Similarly, hepatic uptake of (111)In-GA-BSA after its intravenous injection was not significantly attenuated by co-administration of the anti-CD36 antibody. These results clarify that CD36 does not play a significant role in elimination of GA-BSA and MG-BSA from the circulation, suggesting that the receptor involved in endocytic uptake of circulating AGE proteins by LEC is not SR-A or CD36.


Subject(s)
Acetaldehyde/analogs & derivatives , CD36 Antigens/physiology , Endocytosis , Endothelial Cells/metabolism , Glycation End Products, Advanced/metabolism , Liver/metabolism , Pyruvaldehyde/chemistry , Receptors, Immunologic/physiology , Serum Albumin, Bovine/metabolism , Acetaldehyde/chemistry , Animals , Antibodies, Monoclonal , CD36 Antigens/immunology , Male , Mice , Mice, Knockout , Oleic Acid/metabolism , Rats , Receptors, Scavenger , Scavenger Receptors, Class A , Serum Albumin, Bovine/chemistry
15.
Hypertens Res ; 28(12): 973-80, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16671336

ABSTRACT

Hemodialysis (HD) patients are frequently in a state of increased oxidative stress, and hyperglycemia appears to be a major factor. We recently found that oxidized human serum albumin (HSA) is a reliable marker of oxidative stress in HD patients. However, the issue of whether oxidized HSA is associated with the progression of oxidative stress in HD patients with or without diabetes is not clear. In the present study, we examined the effect of a qualitative modification of HSA in HD patients with or without diabetes. Blood samples from 10 HD patients with diabetes, 7 HD patients without diabetes, and 10 healthy age-matched controls were examined. The increase in plasma protein carbonyl content and advanced glycation endproducts (AGEs) in HD patients was largely due to an increase in the levels of oxidized HSA. Furthermore, these increases were greatest in HD patients with diabetes. Purified HSA from HD patients (non-DM-HSA) was carbonylated and AGE-modified. The amount of modified HSA was the highest in HD patients with diabetes (DM-HSA). Carboxy methyl lysine (CML)-modified HSA triggered a neutrophil respiratory burst, and this activity was closely correlated with the increase in the CML/HSA ratio. These findings indicate that uremia plays an important role in the progression of oxidative stress in HD patients via an increase in CML-modified HSA. They also indicate that diabetic complications further exacerbate the progression of oxidative stress by further increasing the amount of these modified HSA molecules.


Subject(s)
Kidney Failure, Chronic/metabolism , Oxidative Stress/physiology , Renal Dialysis , Serum Albumin/metabolism , Adult , Aged , Aged, 80 and over , Arginine/analogs & derivatives , Arginine/metabolism , Blood Proteins/metabolism , Case-Control Studies , Diabetes Mellitus/metabolism , Diabetes Mellitus/therapy , Female , Glycation End Products, Advanced/metabolism , Humans , Imidazoles/metabolism , Lysine/analogs & derivatives , Lysine/metabolism , Male , Middle Aged , Neutrophils/metabolism , Norleucine/analogs & derivatives , Norleucine/metabolism , Protein Carbonylation , Pyrroles/metabolism , Respiratory Burst
16.
Biochim Biophys Acta ; 1623(2-3): 88-97, 2003 Oct 13.
Article in English | MEDLINE | ID: mdl-14572906

ABSTRACT

Recombinant wild-type human serum albumin (rHSA), the single-residue mutants K199A, K439A and K525A and the triple-residue mutant K199A/K439A/K525A were produced using a yeast expression system. Portions of the rHSA were glycated to different degrees (2.5-250 mM D-glucose). As detected by far-UV and near-UV CD, intrinsic tryptophan-fluorescence and probed by 1,1'-bis(4-anilino)naphthalene-5,5-disulfonic acid, the single-residue mutations had no effect on albumin conformation, whereas the triple-residue mutation and glycation caused conformational changes. The triple-residue mutation and glycation had comparable increased effects on high-affinity binding of warfarin (site I), but decreased effects on high-affinity binding of dansylsarcosine (site II) and the esterase-like activity of albumin. The relation between plasma half-lives in rats were found to be glycated rHSA (50 mM glucose)

Subject(s)
Serum Albumin/chemistry , Serum Albumin/genetics , Animals , Base Sequence , Binding Sites/genetics , Circular Dichroism , DNA, Recombinant/genetics , Esterases/chemistry , Esterases/genetics , Esterases/metabolism , Glycosylation , Humans , In Vitro Techniques , Ligands , Male , Models, Molecular , Mutagenesis, Site-Directed , Mutation , Protein Conformation , Rats , Rats, Wistar , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serum Albumin/metabolism , Spectrometry, Fluorescence
SELECTION OF CITATIONS
SEARCH DETAIL
...