78 results on '"Bijsterbosch MK"'
Search Results
2. Receptor-mediated uptake of low-density lipoprotein by B16 melanoma cells in vitro and in vivo in mice
- Author
-
Versluis, AJ, primary, van Geel, PJ, additional, Oppelaar, H, additional, van Berkel, TJC, additional, and Bijsterbosch, MK, additional
- Published
- 1996
- Full Text
- View/download PDF
3. PLASMA-CLEARANCE AND ENDOCYTOSIS OF MITOCHONDRIAL MALATE-DEHYDROGENASE IN THE RAT
- Author
-
BIJSTERBOSCH, MK, DUURSMA, AM, BOUMA, JMW, GRUBER, M, and NIEUWENHUIS, P
- Published
- 1981
4. Aorta of ApoE-deficient mice responds to atherogenic stimuli by a prelesional increase and subsequent decrease in the expression of antioxidant enzymes.
- Author
-
't Hoen PA, Van der Lans CA, Van Eck M, Bijsterbosch MK, Van Berkel TJ, and Twisk J
- Subjects
- Age Factors, Animals, Aorta pathology, Aorta, Thoracic enzymology, Aorta, Thoracic pathology, Apolipoproteins E genetics, Arteriosclerosis pathology, Catalase genetics, Catalase metabolism, Disease Models, Animal, Disease Progression, Enzymes genetics, Female, Gene Expression Regulation, Glutathione Peroxidase genetics, Glutathione Peroxidase metabolism, Glutathione Transferase genetics, Glutathione Transferase metabolism, Isoenzymes genetics, Isoenzymes metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Oxidative Stress genetics, RNA, Messenger metabolism, Reactive Oxygen Species metabolism, Superoxide Dismutase genetics, Superoxide Dismutase metabolism, Antioxidants metabolism, Aorta enzymology, Apolipoproteins E deficiency, Arteriosclerosis enzymology, Enzymes metabolism
- Abstract
Oxidative stress has been implicated in the development of atherosclerotic lesions. We evaluated the relationship between extent of atherosclerotic lesion formation and vascular expression of pro- and antioxidant enzymes in apoE-deficient mice. On normal chow, these mice showed elevated serum cholesterol levels (7.5- to 9.5-fold), and age-dependent, spontaneous development of all stages of atherosclerotic lesions, starting at the age of 12 weeks. RNA was extracted from the aortic arch and descending aorta, and mRNA expression of pro- and antioxidant enzymes was measured with real-time PCR. Local infiltration of monocytes/macrophages, reflected by increased vascular expression of CD68 mRNA (>10-fold), indicated that the arch was more susceptible than the descending aorta. The expression of catalase-1 and various isoforms of superoxide dismutase, glutathione peroxidase, and glutathione S-transferase alpha was significantly increased in the aortic arch, but not in the descending aorta, in the period preceding lesion formation (age 6 to 12 weeks). These expression levels were 1.5 to 5 times higher than in age-matched wild-type animals. Remarkably, there was an inverse relationship between extent of lesion formation and the mRNA levels of antioxidant enzymes, most of which started to decline after 12 weeks, as lesions developed. In contrast, inducible nitric oxide synthase expression increased 4-fold in the aortic arch over the course of the disease. Our results suggest that the arterial wall responds to increased serum levels of atherogenic lipoproteins by stimulating expression of antioxidant enzymes. The observed co-ordinate decline in expression of many of these protective systems may greatly accelerate the development of atherosclerosis.
- Published
- 2003
- Full Text
- View/download PDF
5. Selection of antisense oligodeoxynucleotides against glutathione S-transferase Mu.
- Author
-
't Hoen PA, Out R, Commandeur JN, Vermeulen NP, van Batenburg FH, Manoharan M, van Berkel TJ, Biessen EA, and Bijsterbosch MK
- Subjects
- Cell-Free System, Dose-Response Relationship, Drug, Glutathione Transferase drug effects, Glutathione Transferase metabolism, Nucleic Acid Conformation, Oligodeoxyribonucleotides, Antisense chemistry, Oligodeoxyribonucleotides, Antisense metabolism, Protein Biosynthesis, Quantitative Structure-Activity Relationship, RNA, Messenger drug effects, RNA, Messenger metabolism, Ribonuclease H metabolism, Software, Transcription, Genetic, Drug Evaluation, Preclinical methods, Glutathione Transferase genetics, Oligodeoxyribonucleotides, Antisense pharmacology, RNA, Messenger chemistry
- Abstract
The aim of the present study was to identify functional antisense oligodeoxynucleotides (ODNs) against the rat glutathione S-transferase Mu (GSTM) isoforms, GSTM1 and GSTM2. These antisense ODNs would enable the study of the physiological consequences of GSTM deficiency. Because it has been suggested that the effectiveness of antisense ODNs is dependent on the secondary mRNA structures of their target sites, we made mRNA secondary structure predictions with two software packages, Mfold and STAR. The two programs produced only marginally similar structures, which can probably be attributed to differences in the algorithms used. The effectiveness of a set of 18 antisense ODNs was evaluated with a cell-free transcription/translation assay, and their activity was correlated with the predicted secondary RNA structures. Four phosphodiester ODNs specific for GSTM1, two ODNs specific for GSTM2, and four ODNs targeted at both GSTM isoforms were found to be potent, sequence-specific, and RNase H-dependent inhibitors of protein expression. The IC50 value of the most potent ODN was approximately 100 nM. Antisense ODNs targeted against regions that were predicted by STAR to be predominantly single stranded were more potent than antisense ODNs against double-stranded regions. Such a correlation was not found for the Mfold prediction. Our data suggest that simulation of the local folding of RNA facilitates the discovery of potent antisense sequences. In conclusion, we selected several promising antisense sequences, which, when synthesized as biologically stable oligonucleotides, can be applied for study of the physiological impact of reduced GSTM expression.
- Published
- 2002
6. bis-Cholesteryl-conjugated phosphorothioate oligodeoxynucleotides are highly selectively taken up by the liver.
- Author
-
Bijsterbosch MK, Manoharan M, Dorland R, Van Veghel R, Biessen EA, and Van Berkel TJ
- Subjects
- Animals, Base Sequence, Biological Transport, Cholesterol blood, Cholesterol pharmacokinetics, Half-Life, Kinetics, Kupffer Cells metabolism, Metabolic Clearance Rate, Mice, Oligodeoxyribonucleotides chemistry, Oligodeoxyribonucleotides, Antisense blood, Oligodeoxyribonucleotides, Antisense chemistry, Organothiophosphorus Compounds chemistry, Rats, Structure-Activity Relationship, Thionucleotides blood, Thionucleotides pharmacokinetics, Tissue Distribution, Cholesterol analogs & derivatives, Liver metabolism, Oligodeoxyribonucleotides pharmacokinetics, Oligodeoxyribonucleotides, Antisense pharmacokinetics, Organothiophosphorus Compounds pharmacokinetics
- Abstract
We previously modulated, by conjugating a single cholesterol, plasma protein binding and liver cell uptake of a phosphorothioate oligodeoxynucleotide (PS-ODN). In this study, we investigated the biological fate of a PS-ODN, denoted ISIS-9389 (3',5'-bis-cholesteryl-conjugated ISIS 3082), provided with two cholesteryl moieties. After intravenous injection of into rats, [(3)H]ISIS-9389 was cleared from plasma with a half-life of 23.6 +/- 0.3 min. After 90 min (approximately 95% cleared), the liver contained 83.0 +/- 0.8% of the dose. Spleen and bone (marrow), which constitute with the liver the reticuloendothelial system, contained 3.1 +/- 0.3 and 4.3 +/- 0.2%, respectively. All other tissues accumulated together <5% of the dose. The hepatic uptake of [(3)H]ISIS-9389 occurred mainly by endothelial cells (51.9 +/- 6.4% of the liver uptake). Parenchymal and Kupffer cells were responsible for 24.9 +/- 7.7 and 23.3 +/- 2.5%, respectively. Preinjected polyinosinic acid and polyadenylic acid reduced hepatic uptake, albeit the latter was less effective. This finding suggests implication of (multiple) scavenger receptors in liver uptake of ISIS-9389. The interaction of ISIS-9389 with plasma proteins, analyzed by size exclusion chromatography, differs from that of unconjugated PS-ODN and PS-ODN with a single cholesterol. Plasma-incubated ISIS-9389 was mainly recovered as a high molecular weight complex. In conclusion, conjugation of PS-ODNs with two cholesteryl moieties results in almost quantitative uptake by the liver. The liver targeting exceeds the already impressive gain in liver uptake achieved by conjugation of a single cholesterol, and is expected to increase the therapeutic activity against liver-associated targets and reduce side effects in nonhepatic tissues.
- Published
- 2002
- Full Text
- View/download PDF
7. Induction of glutathione-S-transferase mRNA levels by chemopreventive selenocysteine Se-conjugates.
- Author
-
't Hoen PA, Rooseboom M, Bijsterbosch MK, van Berkel TJ, Vermeulen NP, and Commandeur JN
- Subjects
- Animals, Cytochrome P-450 Enzyme System biosynthesis, Enzyme Induction drug effects, Hepatocytes drug effects, Hepatocytes metabolism, Lyases metabolism, RNA, Messenger biosynthesis, RNA, Messenger drug effects, Rats, Selenium Compounds metabolism, Selenocysteine chemistry, Tumor Cells, Cultured, Glutathione Transferase biosynthesis, Selenocysteine pharmacology
- Abstract
Several selenocysteine Se-conjugates (SeCys-conjugates) prevent against chemically induced carcinogenesis. Bioactivation to selenols (RSeH) by beta-lyases is thought to be critical, but the mechanism of tumor suppression remains unclear. Induction of phase II biotransformation enzymes is a possible mechanism of chemoprevention. In this study, we evaluated the isoform-selective induction of glutathione-S-transferase (GST) at the mRNA level using a quantitative reverse transcriptase polymerase chain reaction assay. In cultured primary rat hepatocytes and H35 Reuber rat hepatoma cells, SeCys-conjugates time-dependently increased mRNA levels of GST Alpha isoforms and GST Pi, but not of GST Mu isoforms. Se-allyl-L-selenocysteine, the most potent chemopreventive SeCys-conjugate so far known, was also the most active GST inducer. After exposure for 24hr, it elevated GSTA2, GSTA3, GSTA5, and GSTP mRNA levels in primary hepatocytes 3.2+/-0.4-, 1.9+/-0.1-, 4.3+/-0.3-, and 2.9+/-0.3-fold, respectively. Se-allyl-D-selenocysteine was significantly less active, suggesting that stereoselective conversion of SeCys-conjugates to selenols is involved in GST induction. In H35 Reuber hepatoma cells, where conversion of SeCys-conjugates to selenols was 2-6-fold lower than in primary hepatocytes, GST induction was also much lower than in primary hepatocytes. SeCys-conjugates did not induce cytochrome P450 1A1, 2B1/2, or 3A1. This indicates that SeCys-conjugates are monofunctional inducers of phase II biotransformation enzymes. The present results suggest that induction of GST expression contributes to the chemopreventive activity of SeCys-conjugates.
- Published
- 2002
- Full Text
- View/download PDF
8. Selection of effective antisense oligodeoxynucleotides with a green fluorescent protein-based assay. Discovery of selective and potent inhibitors of glutathione S-transferase Mu expression.
- Author
-
't Hoen PA, Rosema BS, Commandeur JN, Vermeulen NP, Manoharan M, van Berkel TJ, Biessen EA, and Bijsterbosch MK
- Subjects
- Animals, COS Cells, Cloning, Molecular, Gene Expression drug effects, Green Fluorescent Proteins, Isoenzymes, Luminescent Proteins genetics, Oligodeoxyribonucleotides, Antisense chemistry, Oligodeoxyribonucleotides, Antisense isolation & purification, Rats, Glutathione Transferase biosynthesis, Oligodeoxyribonucleotides, Antisense pharmacology
- Abstract
Antisense oligodeoxynucleotides (AS-ODNs) are frequently used for the down-regulation of protein expression. Because the majority of potential antisense sequences lacks effectiveness, fast screening methods for the selection of effective AS-ODNs are needed. We describe a new cellular screening assay for the evaluation of the potency and specificity of new antisense sequences. Fusion constructs of the gene of interest and the gene encoding the enhanced green fluorescent protein (EGFP) are cotransfected with AS-ODNs to COS-7 cells. Subsequently, cells are analysed for expression of the EGFP fusion protein by flow cytometry. With the assay, we tested the effectiveness of a set of 15 phosphorothioate ODNs against rat glutathione S-transferase Mu1 (GSTM1) and/or Mu2 (GSTM2). We found several AS-ODNs that demonstrated potent, sequence-specific, and concentration-dependent inhibition of fusion protein expression. At 0.5 microm, AS-6 and AS-8 inhibited EGFP-GSTM1 expression by 95 +/- 4% and 81 +/- 6%, respectively. AS-5 and AS-10 were selective for GSTM2 (82 +/- 4% and 85 +/- 0.4% decrease, respectively). AS-2 and AS-3, targeted at homologous regions in GSTM1 and GSTM2, inhibited both isoforms (77-95% decrease). Other AS-ODNs were not effective or displayed non-target-specific inhibition of protein expression. The observed decrease in EGFP expression was accompanied by a decrease in GSTM enzyme activity. As isoform-selective, chemical inhibitors of GSTM and GSTM knock-out mice are presently unavailable, the selected AS-ODNs constitute important tools for the study of the role of GSTM in detoxification of xenobiotics and protection against chemical-induced carcinogenesis.
- Published
- 2002
- Full Text
- View/download PDF
9. Design of a targeted peptide nucleic acid prodrug to inhibit hepatic human microsomal triglyceride transfer protein expression in hepatocytes.
- Author
-
Biessen EA, Sliedregt-Bol K, 'T Hoen PA, Prince P, Van der Bilt E, Valentijn AR, Meeuwenoord NJ, Princen H, Bijsterbosch MK, Van der Marel GA, Van Boom JH, and Van Berkel TJ
- Subjects
- Animals, Asialoglycoproteins pharmacology, Base Sequence, Biological Transport drug effects, Cells, Cultured, Fetuins, Hepatocytes metabolism, Humans, Kinetics, Mice, Mice, Inbred C57BL, Microsomes, Liver drug effects, Microsomes, Liver metabolism, Molecular Structure, Peptide Nucleic Acids chemistry, Peptide Nucleic Acids genetics, Prodrugs chemistry, Prodrugs pharmacokinetics, RNA, Messenger genetics, RNA, Messenger metabolism, Tumor Cells, Cultured, alpha-Fetoproteins pharmacology, Carrier Proteins genetics, Drug Delivery Systems, Drug Design, Gene Expression Regulation drug effects, Hepatocytes drug effects, Peptide Nucleic Acids chemical synthesis, Peptide Nucleic Acids pharmacology, Prodrugs chemical synthesis, Prodrugs pharmacology
- Abstract
In this study, we present the design and synthesis of an antisense peptide nucleic acid (asPNA) prodrug, which displays an improved biodistribution profile and an equally improved capacity to reduce the levels of target mRNA. The prodrug, K(GalNAc)(2)-asPNA, comprised of a 14-mer sequence complementary to the human microsomal triglyceride transfer protein (huMTP) gene, conjugated to a high-affinity tag for the hepatic asialoglycoprotein receptor (K(GalNAc)(2)). The prodrug was avidly bound and rapidly internalized by HepG2s. After iv injection into mice, K(GalNAc)(2)-asPNA accumulated in the parenchymal liver cells to a much greater extent than nonconjugated PNA (46% +/- 1% vs 3.1% +/- 0.5% of the injected dose, respectively). The prodrug was able to reduce MTP mRNA levels in HepG2 cells by 35-40% (P < 0.02) at 100 nM in an asialoglycoprotein receptor- and sequence-dependent fashion. In conclusion, hepatocyte-targeted PNA prodrugs combine a greatly improved tropism with an enhanced local intracellular availability and activity, making them attractive therapeutics to lower the expression level of hepatic target genes such as MTP.
- Published
- 2002
- Full Text
- View/download PDF
10. Midazolam is a phenobarbital-like cytochrome p450 inducer in rats.
- Author
-
Hoen PA, Bijsterbosch MK, van Berkel TJ, Vermeulen NP, and Commandeur JN
- Subjects
- Animals, Anti-Anxiety Agents pharmacology, Cytochrome P-450 CYP2B6, Cytochrome P-450 CYP3A, Cytochrome P-450 Enzyme System genetics, Cytochrome P-450 Enzyme System metabolism, Enzyme Induction drug effects, Hypnotics and Sedatives pharmacology, Male, Oxidoreductases, N-Demethylating metabolism, RNA, Messenger biosynthesis, RNA, Messenger drug effects, Rats, Rats, Sprague-Dawley, Aryl Hydrocarbon Hydroxylases, Cytochrome P-450 Enzyme System biosynthesis, Midazolam pharmacology, Phenobarbital pharmacology
- Abstract
Midazolam is almost exclusively metabolized by cytochrome P450 3A (CYP3A) isoenzymes. Therefore, midazolam is used as a probe to determine CYP3A levels in humans and rats. A prerequisite for longitudinal determination of CYP3A expression levels using midazolam as a probe is that midazolam itself has no effect on the expression of CYP3A. In the present study, we analyzed the mRNA levels and enzyme activities of the major CYP isoforms in the rat liver after intraperitoneal injection of midazolam (50 mg/kg) for 3 consecutive days. CYP3A1 mRNA levels were increased 4-fold in midazolam-treated animals compared with controls, whereas the mRNA levels of CYP3A2, CYP3A9, and CYP3A18 were not altered. The increase in CYP3A1 mRNA was accompanied by a 25% increase in microsomal testosterone 6beta-hydroxylation activity. More strikingly, CYP2B1/2 mRNA levels were increased 22-fold upon midazolam treatment, leading to an 11- to 95-fold enhancement of CYP2B enzyme activity. CYP2C6 mRNA levels were 4 times higher in midazolam-treated animals. Formation of 2alpha-hydroxy-testosterone, mainly catalyzed by CYP2C11, was 2.6-fold lower in liver microsomes from midazolam-treated animals. Midazolam induced CYP2E enzyme activity 2.5-fold at the post-transcriptional level. The induction of CYP2B1/2 mRNA levels by midazolam was dose-dependent (4.5-fold increase at 10 mg/kg). Induction of CYP3A1 and CYP2B expression was also observed in isolated rat hepatocytes cultured with 100 microM midazolam. We conclude that midazolam is a phenobarbital-like CYP inducer in rats. Induction of CYP3A1 by midazolam may have implications for the longitudinal use of midazolam as a probe for analysis of CYP3A expression levels in rats.
- Published
- 2001
11. Carrier-mediated delivery improves the efficacy of 9-(2-phosphonylmethoxyethyl)adenine against hepatitis B virus.
- Author
-
Bijsterbosch MK, Ying C, de Vrueh RL, de Clercq E, Biessen EA, Neyts J, and van Berkel TJ
- Subjects
- Adenine administration & dosage, Adenine analogs & derivatives, Anti-Inflammatory Agents, Non-Steroidal administration & dosage, Drug Carriers, Hepatitis B virus physiology, Humans, Lactose metabolism, Lipoproteins, HDL, Lithocholic Acid chemistry, Microbial Sensitivity Tests, Tumor Cells, Cultured, Virus Replication drug effects, Adenine pharmacology, Anti-Inflammatory Agents, Non-Steroidal pharmacology, Drug Delivery Systems, Hepatitis B virus drug effects, Organophosphonates
- Abstract
We recently synthesized a lipophilic prodrug of 9-(2-phosphonyl-methoxyethyl)adenine (PMEA), designated PMEA-LO, and incorporated it into reconstituted lactosylated high-density lipoprotein (LacNeoHDL). In a rat model, LacNeoHDL-associated PMEA-LO was internalized by the asialoglycoprotein receptor on parenchymal liver cells and converted into its active diphosphorylated metabolite. To further evaluate the therapeutic potential of the carrier-associated prodrug, we examined in this study the processing of (125)I-labeled PMEA-LO--loaded LacNeoHDL by HepG2 cells. Upon incubation with HepG2 cells, PMEA-LO--loaded LacNeoHDL became rapidly cell-associated. The association was saturable and of high-affinity (k(d) = 3.8 +/- 0.4 nM). Asialofetuin, an established ligand for the asialoglycoprotein receptor, inhibited the association by >75%, which confirms the role of the asialoglycoprotein receptor. Association of the prodrug-loaded particles to HepG2 cells was coupled to degradation. Radiolabeled degradation products appeared in the culture medium with a lag phase of 2 h. Asialofetuin and chloroquine inhibited secretion of degradation products by 75 to 80%, indicating that PMEA-LO--loaded LacNeoHDL is internalized via the asialoglycoprotein receptor and lysosomally processed. The therapeutic potential of LacNeoHDL-associated PMEA-LO was studied by measuring its effects on hepatitis B virus (HBV) replication in Hep AD38 cells (HBV-transfected HepG2 cells). LacNeoHDL-associated PMEA-LO effectively inhibited HBV DNA synthesis. The EC(50) value of carrier-associated PMEA-LO was 35 times lower than that of free PMEA (3.4 +/- 0.4 and 120 +/- 18 ng of PMEA/ml, respectively). We conclude that the present results, combined with our earlier in vivo disposition data, underscore the therapeutic potential and utility of PMEA-LO--loaded LacNeoHDL for treatment of chronic hepatitis B.
- Published
- 2001
12. Delivery of cholesteryl-conjugated phosphorothioate oligodeoxynucleotides to Kupffer cells by lactosylated low-density lipoprotein.
- Author
-
Bijsterbosch MK, Manoharan M, Dorland R, Waarlo IH, Biessen EA, and van Berkel TJ
- Subjects
- Animals, Biological Transport, Cholesterol chemistry, Drug Carriers, Humans, Intercellular Adhesion Molecule-1 drug effects, Lactose chemistry, Liver cytology, Liver metabolism, Male, Rats, Rats, Wistar, Cholesterol administration & dosage, Cholesterol analogs & derivatives, Drug Delivery Systems, Kupffer Cells metabolism, Lipoproteins, LDL chemistry, Oligodeoxyribonucleotides, Antisense administration & dosage, Thionucleotides administration & dosage
- Abstract
The efficacy of antisense oligonucleotides depends on the ability to reach in vivo their target cells. We aim to develop strategies to enhance uptake of phosphorothioate oligodeoxynucleotides by Kupffer cells. To this end, we conjugated cholesterol to ISIS-3082, a phosphorothioate oligodeoxynucleotide specific for intercellular adhesion molecule-1. The cholesterol-conjugated oligonucleotide, denoted ISIS-9388, associated readily with lactosylated low-density lipoprotein (LacLDL), a lipidic carrier that is taken up by galactose receptors on Kupffer cells. Association of up to 10 molecules of ISIS-9388 per LacLDL particle did not induce aggregation. LacLDL-associated [3H]ISIS-9388 was rapidly taken up by the liver after injection into rats (52.9+/-1.8% of the dose within 2 min versus 18.6+/-2.8% for ISIS-3082). N-acetylgalactosamine inhibited hepatic uptake, indicating involvement of galactose-specific receptors. Liver cells were isolated at 60 min after injection of LacLDL-associated [3H]ISIS-9388. Kupffer cells displayed the highest uptake: 88.1+/-24.7 ng of oligonucleotide/mg of cell protein, which is 6-14 times higher than after injection of free ISIS-9388 or ISIS-3082 (15.0+/-3.8 ng and 6.3+/-1.4 ng, respectively). It can be calculated that Kupffer cells contribute 43.9+/-5.4% to the liver uptake (free ISIS-9388 and ISIS-3083 14.5+/-3.1% and 8.3+/-3.2%, respectively). In conclusion, conjugation of a phosphorothioate oligodeoxynucleotide with cholesterol and its subsequent association with LacLDL results in a substantially increased Kupffer cell uptake of the oligonucleotide. As Kupffer cells play a key role in inflammation, our approach may be utilized to improve antisense-based therapeutic intervention during inflammation.
- Published
- 2001
- Full Text
- View/download PDF
13. Recombinant lipoproteins: lipoprotein-like lipid particles for drug targeting.
- Author
-
Rensen PC, de Vrueh RL, Kuiper J, Bijsterbosch MK, Biessen EA, and van Berkel TJ
- Subjects
- Animals, Arteriosclerosis drug therapy, Chylomicrons administration & dosage, Chylomicrons chemistry, Humans, Lipoproteins chemistry, Lipoproteins, HDL administration & dosage, Lipoproteins, HDL chemistry, Lipoproteins, LDL administration & dosage, Lipoproteins, LDL chemistry, Prodrugs chemistry, Recombinant Proteins chemistry, Shock, Septic drug therapy, Surface-Active Agents administration & dosage, Surface-Active Agents chemistry, Tumor Cells, Cultured metabolism, Drug Delivery Systems methods, Hepatocytes metabolism, Lipoproteins administration & dosage, Prodrugs administration & dosage, Recombinant Proteins administration & dosage
- Abstract
Lipoproteins are endogenous particles that transport lipids through the blood to various cell types, where they are recognised and taken up via specific receptors. These particles are, therefore, excellent candidates for the targeted delivery of drugs to various tissues. For example, the remnant receptor and the asialoglycoprotein receptor (ASGPr), which are uniquely localised on hepatocytes, recognise chylomicrons and lactosylated high density lipopoteins (HDL), respectively. In addition, tumour cells of various origins overexpress the low density lipoprotein (LDL) receptor that recognises apolipoprotein E (apoE) on small triglyceride-rich particles and apoB-100 on LDL. Being endogenous, lipoproteins are biodegradable, do not trigger immune reactions, and are not recognised by the reticuloendothelial system (RES). However, their endogenous nature also hampers large-scale pharmaceutical application. In the past two decades, various research groups have successfully synthesised recombinant lipoproteins from commercially available natural and synthetic lipids and serum-derived or recombinant apolipoproteins, which closely mimic the metabolic behaviour of their native counterparts in animal models as well as humans. In this paper, we will summarise the studies that led to the development of these recombinant lipoproteins, and we will address the possibility of using these lipidic particles to selectively deliver a wide range of lipophilic, amphiphilic, and polyanionic compounds to hepatocytes and tumour cells. In addition, the intrinsic therapeutic activities of recombinant chylomicrons and HDL in sepsis and atherosclerosis will be discussed.
- Published
- 2001
- Full Text
- View/download PDF
14. Cryopreservation enables long-term storage of 9-(2-phosphonylmethoxyethyl)adenine prodrug-loaded reconstituted lactosylated high-density lipoprotein.
- Author
-
de Vrueh RL, van Berkel TJ, and Bijsterbosch MK
- Subjects
- Chemical Phenomena, Chemistry, Physical, Cryopreservation, Drug Storage, Electrophoresis, Polyacrylamide Gel, Excipients, Indicators and Reagents, Lactose chemistry, Tissue Distribution, Acrylates chemistry, Lipoproteins, HDL chemistry, Polymers chemistry, Prodrugs chemistry
- Published
- 2001
- Full Text
- View/download PDF
15. Interactions of lipid-oligonucleotide conjugates with low-density lipoprotein.
- Author
-
Rump ET, Biessen EA, van Berkel TJ, and Bijsterbosch MK
- Abstract
The ability of antisense oligonucleotides to interdict, sequence-specifically, the expression of pathogenic genes affords an exciting new strategy for therapeutic intervention (1-3). Oligonucleotides with physiological phosphodiester internucleotide bonds are rapidly degraded, predominantly by exonucleases. Numerous oligonucleotide analogs have therefore been synthesized to confer resistance toward nuclease activity (3). The phosphorothioate analog is the most extensively studied, and phosphorothioate oligodeoxynucleotides have been shown to be potent inhibitors of the expression of their target genes in vitro and in vivo (1,3). However, phosphorothioate oligodeoxynucleotides also bind avidly and nonspecifically to proteins, thus provoking a variety of non-antisense effects (4). Oligonucleotide analogs that do not bind to proteins are therefore expected to display less nonantisense side effects. However, protein binding also affects the in vivo disposition of oligonucleotides. Nonphosphorothioate oligonucelotide analogs generally do not bind to serum proteins, and are therefore rapidly cleared from the circulation, protein-bound phosphorothioate oligodeoxynucelotides circulate much longer (5,6).
- Published
- 2001
- Full Text
- View/download PDF
16. Selective induction of cytochrome P450 3A1 by dexamethasone in cultured rat hepatocytes: analysis with a novel reverse transcriptase-polymerase chain reaction assay section sign.
- Author
-
Hoen PA, Commandeur JN, Vermeulen NP, Van Berkel TJ, and Bijsterbosch MK
- Subjects
- Animals, Anti-Inflammatory Agents pharmacology, Cytochrome P-450 CYP3A, Cytochrome P-450 Enzyme System genetics, Enzyme Induction drug effects, In Vitro Techniques, Liver enzymology, Male, Mixed Function Oxygenases genetics, RNA, Messenger biosynthesis, Rats, Rats, Wistar, Reverse Transcriptase Polymerase Chain Reaction, Testosterone metabolism, Aryl Hydrocarbon Hydroxylases, Cytochrome P-450 Enzyme System biosynthesis, Dexamethasone pharmacology, Liver drug effects, Mixed Function Oxygenases biosynthesis
- Abstract
The study of drug metabolism in cultured rat hepatocytes is hampered by the rapid loss of the expression of cytochrome P450 enzymes. Nevertheless, the activity of cytochrome P450 3A (CYP3A), one of the most important isoenzymes for drug metabolism, can be elevated by chemical inducers. In the present study, we investigated in cultured rat hepatocytes the induction of all four currently identified CYP3A isoforms by dexamethasone, and compared the results obtained in vitro with the induction profile of dexamethasone in vivo. To this end, CYP3A mRNA levels were quantified with a novel, radioactive reverse transcriptase-polymerase chain reaction (RT-PCR) assay, and CYP3A enzymatic activity was measured by a testosterone hydroxylation assay. In the RT-PCR assay, CYP3A isoforms were co-amplified with glyceraldehyde-3-phosphate dehydrogenase (GAPDH) in the presence of radioactively labeled nucleotides. This resulted in an extremely sensitive and accurate determination of CYP3A expression levels, relative to those of GAPDH. Using this RT-PCR assay, it was found that the expression of all CYP3A isoforms in rat hepatocytes, cultured on a collagen matrix, was decreased by 80-90% within one day of cultivation. After addition of dexamethasone, at one day after isolation, CYP3A1 mRNA levels were elevated to levels comparable to those in freshly isolated hepatocytes within two days. In contrast, CYP3A2, CYP3A9, and CYP3A18 mRNA levels were not affected by dexamethasone treatment, and were hardly detectable after three days of cultivation. CYP3A enzymatic activity was also induced in cultured hepatocytes (approximately 6-fold) after addition of dexamethasone. In vivo, CYP3A1 mRNA levels increased 45-fold after dexamethasone administration. However, in contrast to the situation in cultured hepatocytes, CYP3A2 and CYP3A18 were also induced, albeit to a lesser extent (4- and 7-fold elevated mRNA levels, respectively). We conclude that the selective induction of CYP3A1 in dexamethasone-treated rat hepatocytes allows the study of biotransformation reactions by CYP3A1, without interference by any of the other CYP3A isoenzymes.
- Published
- 2000
- Full Text
- View/download PDF
17. Novel hepatotrophic prodrugs of the antiviral nucleoside 9-(2-phosphonylmethoxyethyl)adenine with improved pharmacokinetics and antiviral activity.
- Author
-
Biessen EA, Valentijn AR, De Vrueh RL, Van De Bilt E, Sliedregt LA, Prince P, Bijsterbosch MK, Van Boom JH, Van Der Marel GA, Abrahams PJ, and Van Berkel TJ
- Subjects
- 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, Adenine analogs & derivatives, Adenine pharmacology, Antiviral Agents pharmacology, Herpesvirus 1, Human drug effects, Liver metabolism, Organophosphonates, Prodrugs pharmacology
- 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.
- Published
- 2000
- Full Text
- View/download PDF
18. Modulation of plasma protein binding and in vivo liver cell uptake of phosphorothioate oligodeoxynucleotides by cholesterol conjugation.
- Author
-
Bijsterbosch MK, Rump ET, De Vrueh RL, Dorland R, van Veghel R, Tivel KL, Biessen EA, van Berkel TJ, and Manoharan M
- Subjects
- Animals, Cholesterol blood, Cholesterol chemistry, Liver cytology, Male, Oligodeoxyribonucleotides, Antisense blood, Oligodeoxyribonucleotides, Antisense chemistry, Protein Binding, Rats, Rats, Wistar, Receptors, Immunologic metabolism, Receptors, Scavenger, Scavenger Receptors, Class B, Thionucleotides blood, Thionucleotides chemistry, Tritium, Blood Proteins metabolism, Cholesterol analogs & derivatives, Cholesterol pharmacokinetics, Liver metabolism, Membrane Proteins, Oligodeoxyribonucleotides, Antisense pharmacokinetics, Receptors, Lipoprotein, Thionucleotides pharmacokinetics
- Abstract
Several studies have shown improved efficacy of cholesteryl-conjugated phosphorothioate antisense oligodeoxynucleotides. To gain insight into the mechanisms of the improved efficacy in vivo, we investigated the disposition of ISIS-9388, the 3'-cholesterol analog of the ICAM-1-specific phosphorothioate oligodeoxynucleotide ISIS-3082, in rats. Intravenously injected [(3)H]ISIS-9388 was cleared from the circulation with a half-life of 49.9 +/- 2.2 min (ISIS-3082, 23.3 +/- 3.8 min). At 3 h after injection, the liver contained 63.7 +/- 3. 3% of the dose. Compared to ISIS-3082, the hepatic uptake of ISIS-9388 is approximately 2-fold higher. Endothelial, Kupffer and parenchymal cells accounted for 45.7 +/- 5.7, 33.0 +/- 5.9 and 21.3 +/- 2.6% of the liver uptake of [(3)H]ISIS-9388, respectively, and intracellular concentrations of approximately 2, 75 and 50 microM, respectively, could be reached in these cells (1 mg/kg dose). Preinjection with polyinosinic acid or poly-adenylic acid reduced the hepatic uptake of [(3)H]ISIS-9388, which suggests the involvement of (multiple) scavenger receptors. Size exclusion chromatography of mixtures of the oligonucleotides and rat plasma indicated that ISIS-9388 binds to a larger extent to high molecular weight proteins than ISIS-3082. Analysis by agarose gel electrophoresis indicated that ISIS-9388 binds more tightly to plasma proteins than ISIS-3082. The different interaction of the oligonucleotides with plasma proteins possibly explains their different dispositions. We conclude that cholesterol conjugation results in high accumulation of phosphorothioate oligodeoxynucleotides in various liver cell types, which is likely to be beneficial for antisense therapy of liver-associated diseases.
- Published
- 2000
- Full Text
- View/download PDF
19. Modification of the plasma clearance and liver uptake of steroid ester-conjugated oligodeoxynucleotides by association with (lactosylated) low-density lipoprotein.
- Author
-
Rump ET, de Vrueh RL, Manoharan M, Waarlo IH, van Veghel R, Biessen EA, van Berkel TJ, and Bijsterbosch MK
- Subjects
- Animals, Antineoplastic Agents blood, Humans, Lactose metabolism, Lipoproteins, HDL metabolism, Lipoproteins, LDL blood, Male, Metabolic Clearance Rate, Oligodeoxyribonucleotides, Antisense blood, Rats, Rats, Wistar, Serum Albumin metabolism, Steroids blood, Antineoplastic Agents pharmacokinetics, Lipoproteins, LDL pharmacokinetics, Liver metabolism, Oligodeoxyribonucleotides, Antisense pharmacokinetics, Steroids pharmacokinetics
- Abstract
Low-density lipoprotein (LDL) has been proposed as carrier for the selective delivery of anticancer drugs to tumor cells. We reported earlier the association of several lipidic steroid-conjugated anticancer oligodeoxynucleotides (ODNs) with LDL. In the present study, we determined the stability of these complexes. When the complexes were incubated with a mixture of high-density lipoprotein and albumin, or with rat plasma, the oleoyl steroid-conjugated ODNs appeared to be more stably associated with LDL than the cholesteryl-conjugated ODN. Intravenously injected free lipid-ODNs were very rapidly cleared from the circulation of rats. The area under the curve (AUC) of the lipid-ODNs in plasma was <0.4 microg x min/mL. After complexation with LDL, plasma clearance of the lipid-ODNs was delayed. This was most evident for ODN-5, the ODN conjugated with the oleoyl ester of lithocholic acid (AUC = 6.82 +/- 1.34 microg x min/mL). The AUC of ODN-4, a cholesteryl-conjugated ODN, was 1.49 +/- 0.37 microg x min/mL. In addition, the liver uptake of the LDL-complexed lipid-ODNs was reduced. The lipid-ODNs were also administered as a complex with lactosylated LDL, a modified LDL particle that is selectively taken up by the liver. A high proportion of ODN-5 was transported to the liver along with lactosylated LDL (69.1 +/- 8.1% of the dose at 15 min after injection), whereas much less ODN-4 was transported (36.6 +/- 0.1% of the dose at 15 min after injection). We conclude that the oleoyl ester of lithocholic acid is a more potent lipid anchor than the other steroid lipid anchors. Because of the stable association, the oleoyl ester of lithocholic acid is an interesting candidate for tumor targeting of anticancer ODNs with lipoproteins.
- Published
- 2000
- Full Text
- View/download PDF
20. Carrier-mediated delivery of 9-(2-phosphonylmethoxyethyl)adenine to parenchymal liver cells: a novel therapeutic approach for hepatitis B.
- Author
-
de Vrueh RL, Rump ET, van De Bilt E, van Veghel R, Balzarini J, Biessen EA, van Berkel TJ, and Bijsterbosch MK
- Subjects
- 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, Adenine analogs & derivatives, Antiviral Agents administration & dosage, Antiviral Agents pharmacokinetics, Hepatitis B drug therapy, Liver metabolism, Organophosphonates
- 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.
- Published
- 2000
- Full Text
- View/download PDF
21. Targeted delivery of antisense oligonucleotides to parenchymal liver cells in vivo.
- Author
-
Biessen EA, Vietsch H, Rump ET, Fluiter K, Bijsterbosch MK, and van Berkel TJ
- Subjects
- Animals, Asialoglycoproteins metabolism, Biological Availability, Drug Carriers, Drug Stability, Glycopeptides, Liver cytology, Oligodeoxyribonucleotides, Antisense chemistry, Oligodeoxyribonucleotides, Antisense pharmacokinetics, Orosomucoid metabolism, Rats, Receptors, Scavenger, Scavenger Receptors, Class B, Drug Delivery Systems methods, Liver drug effects, Membrane Proteins, Oligodeoxyribonucleotides, Antisense administration & dosage, Receptors, Immunologic metabolism, Receptors, Lipoprotein
- Published
- 2000
- Full Text
- View/download PDF
22. Synthesis of a lipophilic prodrug of 9-(2-phosphonylmethoxyethyl)adenine (PMEA) and its incorporation into a hepatocyte-specific lipidic carrier.
- Author
-
de Vrueh RL, Rump ET, Sliedregt LA, Biessen EA, van Berkel TJ, and Bijsterbosch MK
- Subjects
- Adenine administration & dosage, Adenine pharmacology, Animals, Antiviral Agents pharmacology, Drug Carriers, Drug Delivery Systems, Humans, Lipoproteins, HDL metabolism, Lithocholic Acid metabolism, Lithocholic Acid pharmacology, Liver drug effects, Prodrugs metabolism, Prodrugs pharmacology, Rats, Tritium, Adenine analogs & derivatives, Antiviral Agents administration & dosage, Lithocholic Acid analogs & derivatives, Lithocholic Acid chemical synthesis, Liver metabolism, Organophosphonates, Prodrugs chemical synthesis
- Abstract
Purpose: 9-(2-Phosphonylmethoxyethyl)adenine (PMEA), a potent inhibitor of Hepatitis B virus replication, is in vivo hardly taken up by parenchymal liver cells (the site of infection). Our aim is to examine whether lactosylated reconstituted HDL (LacNeoHDL), a lipidic particle that is specifically internalized by parenchymal liver cells, is a suitable carrier for the selective delivery of PMEA to this cell type., Methods: To incorporate PMEA into LacNeoHDL, we synthesized a lipophilic prodrug (PMEA-LO) by coupling PMEA via an acid-labile phosphonamidate bond to lithocholic acid-3alpha-oleate., Results: The yield of the synthesis was 52% ([3H]PMEA-LO: 24%). [3H]PMEA-LO readily incorporated into LacNeoHDL (13 molecules/particle) without affecting the size and net negative charge of the carrier. Further, incubation studies at lysosomal pH showed [3H]PMEA was completely released from the carrier whereas, at neutral pH or in plasma, appreciable release was not observed., Conclusions: The conjugation of PMEA with lithocholic acid-3alpha-oleate results in a lipophilic prodrug that readily associates with Lac-NeoHDL. The association of the prodrug does not affect the physicochemical properties of the particle, and PMEA is released from the carrier at lysosomal pH. These findings indicate that by using the prodrug approach, LacNeoHDL is a suitable carrier to deliver PMEA to parenchymal liver cells.
- Published
- 1999
- Full Text
- View/download PDF
23. Targeted delivery of oligodeoxynucleotides to parenchymal liver cells in vivo.
- Author
-
Biessen EA, Vietsch H, Rump ET, Fluiter K, Kuiper J, Bijsterbosch MK, and van Berkel TJ
- Subjects
- Acetylglucosamine metabolism, Acetylglucosamine pharmacology, Animals, Asialoglycoprotein Receptor, Binding, Competitive, Cells, Cultured, Colchicine pharmacology, Endocytosis drug effects, Galactosides chemical synthesis, Galactosides chemistry, Galactosides isolation & purification, Galactosides metabolism, Gene Expression Regulation drug effects, Half-Life, Ligands, Lysosomes metabolism, Male, Microscopy, Confocal, Monensin pharmacology, Oligodeoxyribonucleotides, Antisense blood, Oligodeoxyribonucleotides, Antisense chemical synthesis, Oligodeoxyribonucleotides, Antisense genetics, Rats, Rats, Wistar, Receptors, Cell Surface metabolism, Sodium Azide pharmacology, Sucrose pharmacology, Gene Targeting, Liver cytology, Liver metabolism, Oligodeoxyribonucleotides, Antisense metabolism
- Abstract
Anti-sense oligodeoxynucleotides (ODNs) hold great promise for correcting the biosynthesis of clinically relevant proteins. The potential of ODNs for modulating liver-specific genes might be increased by preventing untimely elimination and by improving the local bioavailability of ODNs in the target tissue. In the present study we have assessed whether the local ODN concentration can be enhanced by the targeted delivery of ODNs through conjugation to a ligand for the parenchymal liver cell-specific asialoglycoprotein receptor. A capped ODN (miscellaneous 20-mer sequence) was derivatized with a ligand with high affinity for this receptor, N2-[N2-(N2,N6-bis{N-[p-(beta-d-galactopyranosyloxy) anilino] thiocarbamyl}-L-lysyl)-N6-(N-{p-[beta-D -galactopyranosyloxy] anilino} thiocarbamyl)-L-lysyl]-N6-[N- (p-{beta-D-galactopyranosyloxy}anilino)thiocarbamyl]-L-lysine (L3G4) (Kd 6.5+/-0.2 nM, mean+/-S.D.). Both the uptake studies in vitro and the confocal laser scan microscopy studies demonstrated that L3G4-ODN was far more efficiently bound to and taken up by parenchymal liver cells than underivatized ODN. Studies in vivo in rats showed that hepatic uptake could be greatly enhanced from 19+/-1% to 77+/-6% of the injected dose after glycoconjugation. Importantly, specific ODN accumulation of ODN into parenchymal liver cells was improved almost 60-fold after derivatization with L3G4, and could be attributed to the asialoglycoprotein receptor. In conclusion, the scavenger receptor-mediated elimination pathway for miscellaneous ODN sequences can be circumvented by direct conjugation to a synthetic tag for the asialoglycoprotein receptor. In this manner a crucial requisite is met towards the application of ODNs in vivo to modulate the biosynthesis of parenchymal liver cell-specific genes such as those for apolipoprotein (a), cholesterol ester transfer protein and viral proteins.
- Published
- 1999
24. Stable incorporation of a lipophilic daunorubicin prodrug into apolipoprotein E-exposing liposomes induces uptake of prodrug via low-density lipoprotein receptor in vivo.
- Author
-
Versluis AJ, Rump ET, Rensen PC, van Berkel TJ, and Bijsterbosch MK
- Subjects
- Animals, Antibiotics, Antineoplastic administration & dosage, Antibiotics, Antineoplastic blood, Antibiotics, Antineoplastic chemistry, Apolipoproteins E, Daunorubicin administration & dosage, Daunorubicin blood, Daunorubicin chemistry, Drug Carriers, Ethinyl Estradiol pharmacology, Humans, Iodine Radioisotopes, Liposomes, Liver metabolism, Male, Mice, Mice, Inbred C57BL, Prodrugs administration & dosage, Prodrugs chemistry, Rats, Rats, Wistar, Receptors, LDL deficiency, Receptors, LDL genetics, Recombinant Proteins metabolism, Antibiotics, Antineoplastic pharmacokinetics, Daunorubicin pharmacokinetics, Prodrugs pharmacokinetics, Receptors, LDL metabolism
- Abstract
Many tumors express elevated levels of low-density lipoprotein (LDL) receptors. Therefore, native LDL and synthetic LDL-like particles have been proposed as carriers for antineoplastic drugs. We demonstrated earlier that small apolipoprotein E (apoE)-exposing liposomes were specifically recognized by the LDL receptor. In this study, we incorporated a lipophilic derivative of daunorubicin (LAD) into the apoE liposomes. Up to 11 molecules of LAD could be incorporated per particle without significantly changing the size, lipid composition, and ability to bind apoE of the liposomes. The biological fate of the prodrug was largely determined by its carrier (70% of the initially incorporated LAD was still associated to the liposomes after 4 h of circulation in mice). Compared with free daunorubicin, the circulation half-life of the liposome-associated prodrug was substantially prolonged and undesired tissue disposition was reduced. The role of the LDL receptor in the metabolism of LAD-loaded apoE liposomes was demonstrated in rats with up-regulated hepatic LDL receptors. In these rats, the liver uptake of the prodrug and carrier was increased 5-fold. The addition of apoE was essential for LDL receptor-mediated uptake of the drug-carrier complex. In LDL receptor-deficient mice, the circulation time of both the prodrug and the carrier increased approximately 2-fold compared with wild-type mice. We conclude that LAD-loaded apoE liposomes constitute a stable drug-carrier complex that is well suited for LDL receptor-mediated selective drug delivery to tumors.
- Published
- 1999
25. Design and synthesis of novel amphiphilic dendritic galactosides for selective targeting of liposomes to the hepatic asialoglycoprotein receptor.
- Author
-
Sliedregt LA, Rensen PC, Rump ET, van Santbrink PJ, Bijsterbosch MK, Valentijn AR, van der Marel GA, van Boom JH, van Berkel TJ, and Biessen EA
- Subjects
- Animals, Asialoglycoprotein Receptor, Binding, Competitive, Drug Design, Galactosides chemistry, Glycolipids chemistry, In Vitro Techniques, Liposomes metabolism, Liver cytology, Liver metabolism, Male, Mice, Mice, Inbred C57BL, Rats, Structure-Activity Relationship, Asialoglycoproteins metabolism, Galactosides chemical synthesis, Glycolipids chemical synthesis, Liposomes chemistry, Receptors, Cell Surface drug effects
- Abstract
A series of glycolipids have been prepared which contain a cluster galactoside moiety with high affinity for the hepatic asialoglycoprotein receptor and a bile acid ester moiety which mediates stable incorporation into liposomes. Loading of liposomes with these glycolipids at a ratio of 5% (w/w) resulted in efficient recognition and uptake of the liposomes by the liver. Preinjection with asialofetuin almost completely inhibited the uptake, establishing that the liposomes were selectively recognized and processed by the asialoglycoprotein receptor on liver parenchymal cells. In contrast, a glycolipid content of 50% (w/w) led to a liver uptake that could not be inhibited by preinjection with asialofetuin, indicating that the liposomes were now processed by the Gal/Fuc-recognizing receptor on liver macrophages. The results presented in this study are important for future targeting of water-soluble and amphiphilic drugs, enveloped in these glycolipid-laden liposomes, to parenchymal liver cells.
- Published
- 1999
- Full Text
- View/download PDF
26. Preparation of conjugates of oligodeoxynucleotides and lipid structures and their interaction with low-density lipoprotein.
- Author
-
Rump ET, de Vrueh RL, Sliedregt LA, Biessen EA, van Berkel TJ, and Bijsterbosch MK
- Subjects
- Bile Acids and Salts chemistry, Cholesterol analogs & derivatives, Electrophoresis, Agar Gel, Molecular Structure, Oleic Acids chemistry, Oligonucleotides, Antisense genetics, Proto-Oncogene Proteins chemistry, Proto-Oncogene Proteins c-myb, Steroids chemistry, Trans-Activators chemistry, Lipids chemistry, Lipoproteins, LDL metabolism, Oligodeoxyribonucleotides chemistry
- Abstract
The high expression level of receptors for low-density lipoprotein (LDL) on tumor cells makes LDL an attractive carrier for selective delivery of drugs to these cells. The aim of this study is to allow incorporation of oncogene-directed antisense oligodeoxynucleotides (ODNs) into the lipid moiety of LDL. Therefore, ODNs were conjugated with oleic acid, cholesterol, and several other steroid lipids. These latter steroid lipids were synthesized starting from bile acids and were varied in lipophilicity by attaching oleic acid ester structures. The lipid structures, activated as pentafluorophenyl esters, were conjugated in solution phase to 3'-amino-tailed ODNs. The ODNs conjugated with lithocholic acid, oleic acid, and cholesterol could easily be purified by reversed phase (RP)-HPLC. However, the ODNs conjugated with the oleoyl steroid ester structures irreversibly bound to the column material. These highly lipidic ODNs were separated from the nonconjugated ODN by electrophoresis in a 1% low-melting agarose gel containing 0.1% Tween 20. This method was found to be very effective in isolating the ODNs conjugated to the oleoyl steroid ester structures. The ODNs conjugated with cholesterol and the oleoyl esters of lithocholic and cholenic acid associated readily and nearly completely with LDL. However, the less lipidic ODNs and the ODN conjugated with the dioleoyl ester of chenodeoxycholic acid did not and did incompletely associate, respectively. Lithocholic acid and oleic acid are probably not sufficiently lipophilic to induce association with LDL, whereas the dioleoyl ester structure is probably too bulky and extended to allow partitioning into the lipid moiety of LDL. We conclude that several of the lipid-ODNs can associate readily with LDL, enabling delivery of oncogene-directed antisense ODNs via the LDL receptor pathway.
- Published
- 1998
- Full Text
- View/download PDF
27. Disposition of the acyclic nucleoside phosphonate (S)-9-(3-hydroxy-2-phosphonylmethoxypropyl)adenine.
- Author
-
Bijsterbosch MK, Smeijsters LJ, and van Berkel TJ
- Subjects
- Adenine blood, Adenine pharmacokinetics, Animals, Antiviral Agents blood, Chromatography, High Pressure Liquid, Kidney metabolism, Male, Metabolic Clearance Rate, Organophosphorus Compounds blood, Radiopharmaceuticals blood, Rats, Rats, Wistar, Tissue Distribution, Adenine analogs & derivatives, Antiviral Agents pharmacokinetics, Liver metabolism, Organophosphonates, Organophosphorus Compounds pharmacokinetics, Radiopharmaceuticals pharmacokinetics
- Abstract
The acyclic nucleoside phosphonate (S)-9-(3-hydroxy-2-phosphonylmethoxypropyl)adenine [(S)-HPMPA] has been shown to be active against pathogens, like hepatitis B viruses and Plasmodium parasites, that infect parenchymal liver cells. (S)-HPMPA is therefore an interesting candidate drug for the treatment of these infections. To establish effective therapeutic protocols for (S)-HPMPA, it is essential that the kinetics of its hepatic uptake be evaluated and that the role of the various liver cell types be examined. In the present study, we investigated the disposition of (S)-HPMPA and assessed its hepatic uptake. Rats were intravenously injected with [3H](S)-HPMPA, and after an initial rapid distribution phase (360 +/- 53 ml/kg of body weight), the radioactivity was cleared from the circulation with a half-life of 11.7 +/- 1.4 min. The tissue distribution of [3H](S)-HPMPA was determined at 90 min after injection (when >99% of the dose cleared). Most (57.0% +/- 1.1%) of the injected [3H](S)-HPMPA was excreted unchanged in the urine. The radioactivity that was retained in the body was almost completely recovered in the kidneys and the liver (68.4% +/- 2.5% and 16.1% +/- 0.4% of the radioactivity in the body, respectively). The uptake of [3H](S)-HPMPA by the liver occurred mainly by parenchymal cells (92.1% +/- 3.4% of total uptake by the liver). Kupffer cells and endothelial cells accounted for only 6.1% +/- 3.5% and 1.8% +/- 0.8% of the total uptake by the liver, respectively. Preinjection with probenecid reduced the hepatic and renal uptake of [3H](S)-HPMPA by approximately 75%, which points to a major role of a probenecid-sensitive transporter in the uptake of (S)-HPMPA by both tissues. In conclusion, we show that inside the liver, (S)-HPMPA is mainly taken up by parenchymal liver cells. However, the level of uptake by the kidneys is much higher, which leads to nephrotoxicity. An approach in which (S)-HPMPA is coupled to carriers that are specifically taken up by parenchymal cells may increase the effectiveness of the drug in the liver and reduce its renal toxicity.
- Published
- 1998
- Full Text
- View/download PDF
28. Synthesis of a lipophilic daunorubicin derivative and its incorporation into lipidic carriers developed for LDL receptor-mediated tumor therapy.
- Author
-
Versluis AJ, Rump ET, Rensen PC, Van Berkel TJ, and Bijsterbosch MK
- Subjects
- Antibiotics, Antineoplastic blood, Cholesterol Esters chemistry, Cholic Acids metabolism, Daunorubicin analogs & derivatives, Daunorubicin metabolism, Drug Carriers chemistry, Emulsions, Humans, Liposomes, Oligopeptides chemistry, Structure-Activity Relationship, Antibiotics, Antineoplastic chemical synthesis, Cholic Acids chemical synthesis, Daunorubicin chemical synthesis, Prodrugs chemical synthesis, Receptors, LDL metabolism
- Abstract
Purpose: Many tumors express elevated levels of LDL receptors (apoB, E receptors) on their membranes. Selective delivery of anti-neoplastic drugs to tumors by incorporation of these drugs into LDL or LDL-resembling particles should improve the efficacy of tumor therapy and minimize the severe side-effects. Since the apolipoproteins on the particles are essential for the LDL receptor recognition, drugs should preferably be incorporated into the lipid moiety. Most anti-tumor agents are too hydrophilic for incorporation into these carriers., Methods: We synthesized LAD, a lipophilic prodrug of daunorubicin, by coupling the drug via a lysosomally degradable peptide spacer to a cholesteryl oleate analog., Results: The overall yield of the synthesis was 50% with a purity of > 90%. Radioactively ([3H]) labeled LAD was obtained via a slightly modified procedure (yield 40%). The octanol/water partition coefficient of LAD is 30-fold higher than that of daunorubicin. LAD could be incorporated into triglyceride-rich lipid emulsions and small liposomes, which, if provided with apoE, have been demonstrated earlier to be cleared in vivo via the LDL receptor. The liposomes contained approximately 10 molecules of LAD per liposomal particle. Analysis of differently sized LAD-containing emulsions suggests that LAD associates with the surface of lipidic particles. In the presence of human serum, LAD did not dissociate from the emulsion particles, indicating a firm association of LAD with the carrier., Conclusions: The coupling of a cholesterol ester analog to daunorubicin results in a lipophilic prodrug that can be firmly anchored into lipidic carries. LAD-loaded emulsions and liposomes provided with recombinant apoE will be tested in the near future for their ability to deliver LAD to tumor tissue in vivo via the LDL receptor.
- Published
- 1998
- Full Text
- View/download PDF
29. Liver uptake of phosphodiester oligodeoxynucleotides is mediated by scavenger receptors.
- Author
-
Biessen EA, Vietsch H, Kuiper J, Bijsterbosch MK, and Berkel TJ
- Subjects
- Animals, Biological Transport, Endothelium metabolism, Kupffer Cells metabolism, Lipoproteins, LDL metabolism, Male, Metabolic Clearance Rate, Polyelectrolytes, Polymers metabolism, Rats, Rats, Wistar, Receptors, Scavenger, Scavenger Receptors, Class B, Liver metabolism, Membrane Proteins, Oligonucleotides, Antisense metabolism, Receptors, Immunologic metabolism, Receptors, Lipoprotein
- Abstract
The therapeutic activity of antisense oligodeoxynucleotides (ODNs) often is impaired due to premature degradation and poor ability to reach the (intra)cellular target. In this study, we addressed the in vivo fate of ODNs and characterized the major sites responsible for the clearance of intravenously injected phosphodiester ODN. On injection into rats, 32P-ODNs (miscellaneous sequences and GT-containing ODNs with variable G content) are rapidly cleared from the bloodstream (t1/2 = 0.6-0.7 min), with the liver being the main site of elimination. The contribution of the liver to ODN clearance depended on its sequence and varied considerably. Hepatic uptake tended to be lower for G-rich ODNs as a result of increased bone marrow uptake. Within the liver, both Kupffer cells (KC) and endothelial cells (EC) were responsible for 32P-ODN uptake. To elucidate the mechanism of liver uptake, 32P-ODN binding studies using isolated EC and KC were performed. Binding to both cell types seemed to be saturable, of moderate affinity, and mediated by a membrane-bound protein. The inhibition profiles of 32P-ODN binding to EC and KC by various (poly)anions were essentially equal and corresponded closely to those of 125I-acetylated low-density lipoprotein. In summary, the results indicate that scavenger receptors on nonparenchymal liver and bone marrow cells contribute to the elimination of ODNs from the bloodstream. Minor changes in ODN sequence markedly affect receptor recognition, resulting in considerable shifts in the biodistribution of antisense ODNs.
- Published
- 1998
- Full Text
- View/download PDF
30. Human recombinant apolipoprotein E-enriched liposomes can mimic low-density lipoproteins as carriers for the site-specific delivery of antitumor agents.
- Author
-
Rensen PC, Schiffelers RM, Versluis AJ, Bijsterbosch MK, Van Kuijk-Meuwissen ME, and Van Berkel TJ
- Subjects
- Animals, Antineoplastic Agents pharmacokinetics, Apolipoproteins E administration & dosage, Drug Carriers, Humans, Lipoproteins, LDL pharmacokinetics, Liver metabolism, Male, Melanoma, Experimental metabolism, Rats, Rats, Wistar, Receptors, LDL metabolism, Recombinant Proteins chemical synthesis, Recombinant Proteins metabolism, Recombinant Proteins pharmacology, Sensitivity and Specificity, Antineoplastic Agents administration & dosage, Apolipoproteins E chemical synthesis, Apolipoproteins E metabolism, Lipoproteins, LDL administration & dosage, Liposomes chemical synthesis, Liposomes metabolism
- Abstract
Progressive hypocholesterolemia is a feature associated with a number of cancers of different origin, and it is caused by the high expression of low-density lipoprotein (LDL) receptors (LDLrs) on many tumor cell types. Selective delivery of chemotherapeutics using LDL as a carrier has therefore been proposed, but the endogenous nature of LDL hampers its pharmaceutical application. In the current study, we explored the possibility of synthesizing liposomes that mimic LDL from commercially available lipids and proteins. Small unilamellar liposomes were created (28.9 +/- 0.9 nm) and complexed with 5.8 +/- 0.7 molecules of human recombinant apolipoprotein E (apoE). On intravenous injection into rats, the liposomes retained their aqueous core, structural integrity, and the majority of the preassociated apoE. [3H]Cholesteryl oleate-labeled apoE-enriched liposomes showed a relatively long serum half-life (>5 hr), and a low uptake by cells of the reticuloendothelial system was observed (<0.8% of the injected dose at 30 min after injection). Pretreatment of rats with 17alpha-ethinyl estradiol, which induces the expression of the LDLr on the liver and adrenals, led to a 2.5-fold accelerated serum clearance (t1/2 = 123 +/- 10 min) and a selectively increased uptake of liposomes by the liver (2.0-fold) and adrenals (3.8-fold). The liver association of the liposomes was coupled to the lysosomal uptake route, similarly as for LDL. In vitro studies using B16 melanoma cells showed that the liposomes bound exclusively to the LDLr via their apoE moiety (90,000 liposomes/cell), with a 14-fold higher affinity (Kd = 0.77 +/- 0.09 nM) than LDL itself. Because of their favorable properties, we anticipate that these apoE-enriched liposomes are advantageous compared with native LDL in the development of a selective LDLr-targeted antitumor therapy.
- Published
- 1997
- Full Text
- View/download PDF
31. In vivo fate of phosphorothioate antisense oligodeoxynucleotides: predominant uptake by scavenger receptors on endothelial liver cells.
- Author
-
Bijsterbosch MK, Manoharan M, Rump ET, De Vrueh RL, van Veghel R, Tivel KL, Biessen EA, Bennett CF, Cook PD, and van Berkel TJ
- Subjects
- Animals, Endothelium metabolism, Intercellular Adhesion Molecule-1 genetics, Male, Metabolic Clearance Rate, Mice, Polyelectrolytes, Polymers metabolism, Rats, Rats, Wistar, Receptors, Scavenger, Scavenger Receptors, Class B, Subcellular Fractions metabolism, Tissue Distribution, Liver metabolism, Membrane Proteins, Oligonucleotides, Antisense metabolism, Receptors, Immunologic metabolism, Receptors, Lipoprotein, Thionucleotides metabolism
- Abstract
Systemically administered phosphorothioate antisense oligodeoxynucleotides can specifically affect the expression of their target genes, which affords an exciting new strategy for therapeutic intervention. Earlier studies point to a major role of the liver in the disposition of these oligonucleotides. The aim of the present study was to identify the cell type(s) responsible for the liver uptake of phosphorothioate oligodeoxynucleotides and to examine the mechanisms involved. In our study we used ISIS-3082, a phosphorothioate antisense oligodeoxynucleotide specific for murine ICAM-1. Intravenously injected [3H]ISIS-3082 (dose: 1 mg/kg) was cleared from the circulation of rats with a half-life of 23.3+/-3.8 min. At 90 min after injection (>90% of [3H]ISIS-3082 cleared), the liver contained the most radioactivity, whereas the second-highest amount was recovered in the kidneys (40.5+/-1.4% and 17.9+/-1.3% of the dose, respectively). Of the remaining tissues, only spleen and bone marrow actively accumulated [3H]ISIS-3082. By injecting different doses of [3H]ISIS-3082, it was found that uptake by liver, spleen, bone marrow, and kidneys is saturable, which points to a receptor-mediated process. Subcellular fractionation of the liver indicates that ISIS-3082 is internalized and delivered to the lysosomes. Liver uptake occurs mainly (for 56.1+/-3.0%) by endothelial cells, whereas parenchymal and Kupffer cells account for 39.6+/-4.5 and 4.3+/-1.7% of the total liver uptake, respectively. Preinjection of polyinosinic acid substantially reduced uptake by liver and bone marrow, whereas polyadenylic acid was ineffective, which indicates that in these tissues scavenger receptors are involved in uptake. Polyadenylic acid, but not polyinosinic acid, reduced uptake by kidneys, which suggests renal uptake by scavenger receptors different from those in the liver. We conclude that scavenger receptors on rat liver endothelial cells play a predominant role in the plasma clearance of ISIS-3082. As scavenger receptors are also expressed on human endothelial liver cells, our findings are probably highly relevant for the therapeutic application of phosphorothioate oligodeoxynucleotides in humans. If the target gene is not localized in endothelial liver cells, the therapeutic effectiveness might be improved by developing delivery strategies that redirect the oligonucleotides to the actual target cells.
- Published
- 1997
- Full Text
- View/download PDF
32. Antagonists of the mannose receptor and the LDL receptor-related protein dramatically delay the clearance of tissue plasminogen activator.
- Author
-
Biessen EA, van Teijlingen M, Vietsch H, Barrett-Bergshoeff MM, Bijsterbosch MK, Rijken DC, van Berkel TJ, and Kuiper J
- Subjects
- Animals, Cells, Cultured, Endocytosis drug effects, Half-Life, Humans, Liver metabolism, Low Density Lipoprotein Receptor-Related Protein-1, Mannose Receptor, Mannosides toxicity, Oligopeptides toxicity, Rats, Rats, Wistar, Receptors, Cell Surface metabolism, Receptors, Immunologic metabolism, Receptors, LDL antagonists & inhibitors, Receptors, LDL metabolism, Recombinant Fusion Proteins pharmacology, Carrier Proteins pharmacology, Glutathione Transferase pharmacology, Lectins, C-Type, Mannose-Binding Lectins, Mannosides pharmacology, Oligopeptides pharmacology, Receptors, Cell Surface antagonists & inhibitors, Receptors, Immunologic antagonists & inhibitors, Tissue Plasminogen Activator pharmacokinetics
- Abstract
Background: Clinical application of tissue plasminogen activator (TPA) as a fibrinolytic agent is complicated by its rapid clearance from the bloodstream, which is caused by TPA liver uptake. The mannose receptor on endothelial liver cells and the LDL receptor-related protein (LRP) on parenchymal liver cells were reported to contribute to liver uptake., Methods and Results: In this study, we addressed whether TPA clearance can be delayed by inhibiting receptor-mediated endocytosis of TPA. A series of cluster mannosides was synthesized, and their affinity for the mannose receptor was determined. A cluster mannoside carrying six mannose groups (M6L5) displayed a subnanomolar affinity for the mannose receptor (Ki = 0.41 +/- 0.09 nmol/L). Preinjection of M6L5 (1.2 mg/kg) reduced the clearance of 125I-TPA in rats by 60% because of specific inhibition of the endothelial cell uptake. The low toxicity of M6L5, combined with its accessible synthesis and high specificity for the mannose receptor, makes it a promising agent to improve the pharmacokinetics of TPA. Blockade of LRP by 39-kD receptor-associated protein (GST-RAP) also inhibited TPA clearance by 60%. Finally, combined preinjection of M6L5 and GST-RAP almost completely abolished reduced liver uptake of TPA and delayed its clearance by a factor of 10., Conclusions: It can be concluded that (1) the mannose receptor and LRP appear to be the sole major receptors responsible for TPA clearance and (2) therapeutic levels of TPA can be maintained for a prolonged time span by coadministration of the aforementioned receptor antagonists.
- Published
- 1997
- Full Text
- View/download PDF
33. Lysine-based cluster mannosides that inhibit ligand binding to the human mannose receptor at nanomolar concentration.
- Author
-
Biessen EA, Noorman F, van Teijlingen ME, Kuiper J, Barrett-Bergshoeff M, Bijsterbosch MK, Rijken DC, and van Berkel TJ
- Subjects
- Binding, Competitive, Biotin, Chromatography, Thin Layer, Humans, Kinetics, Magnetic Resonance Spectroscopy, Mannose Receptor, Oligosaccharides metabolism, Ribonucleases metabolism, Tissue Plasminogen Activator pharmacology, Lectins metabolism, Lectins, C-Type, Lysine, Mannose-Binding Lectins, Oligosaccharides chemical synthesis, Receptors, Cell Surface metabolism
- Abstract
In search of synthetic high affinity ligands for the mannose receptor, we synthesized a series of lysine-based oligomannosides containing two (M2L) to six (M6L5) terminal alpha-D-mannose groups that are connected with the backbone by flexible elongated spacers (16 A). The synthesized cluster mannosides were all able to displace binding of biotinylated ribonuclease B and tissue-type plasminogen activator to isolated human mannose receptor. The affinity of these cluster mannosides for the mannose receptor was continuously enhanced from 18-23 microM to 0.5-2.6 nM, with mannose valencies increasing from two to six. On average, expansion of the cluster mannoside with an additional alpha-D-mannose group resulted in a 10-fold increase in its affinity for the mannose receptor. M3L2 to M6L5 displayed negative cooperative inhibition of ligand binding to the mannose receptor, suggesting that binding of these mannosides involves multiple binding sites. The nanomolar affinity of the most potent ligand, the hexamannoside M6L5 makes it the most potent synthetic cluster mannoside for the mannose receptor yet developed. As a result of its high affinity and accessible synthesis, M6L5 not only is a powerful tool to study the mechanism of ligand binding by the mannose receptor, but it is also a promising targeting device to accomplish cell-specific delivery of genes and drugs to liver endothelial cells or macrophages in bone marrow, lungs, spleen, and atherosclerotic plaques.
- Published
- 1996
- Full Text
- View/download PDF
34. Specific targeting of a lipophilic prodrug of iododeoxyuridine to parenchymal liver cells using lactosylated reconstituted high density lipoprotein particles.
- Author
-
Bijsterbosch MK, van de Bilt H, and van Berkel TJ
- Subjects
- Animals, Antiviral Agents administration & dosage, Drug Carriers, Idoxuridine administration & dosage, Lactose chemistry, Lipoproteins, HDL chemistry, Prodrugs administration & dosage, Rats, Subcellular Fractions metabolism, Tritium, Antiviral Agents pharmacokinetics, Idoxuridine pharmacokinetics, Lipoproteins, HDL administration & dosage, Liver metabolism, Prodrugs pharmacokinetics
- Abstract
We recently reported the conversion of the water-soluble antiviral drug iododeoxyuridine (IDU) into the lipophilic prodrug dioleoyl-iododeoxyuridine (IDU-Ol2). The prodrug was incorporated into reconstituted high-density lipoprotein (NeoHDL) particles with physical and biological properties similar to those of native HDL. We also found, in initial experiments, that lactosylation of the prodrug-loaded NeoHDL increases its liver uptake. Because this offers the attractive perspective of using these particles for the delivery of drugs to the liver, we now analyze the characteristics and biological fate of lactosylated IDU-Ol2-loaded NeoHDL. The particles (containing approximately 25 prodrug molecules) have the same size and charge as native HDL, indicating that lactosylation does not cause aggregation or oxidative modification. At 10 min after intravenous injection of lactosylated [3H]IDU-Ol2-loaded NeòHDL into rats, only 13.5 +/- 2.8% of the dose was left in plasma and 75.9 +/- 2.4% of the dose was recovered in the liver. The relative specific uptake by the liver was 1-2 orders of magnitude higher than that of any other tissue. The hepatic uptake of lactosylated [3H]IDU-Ol2-loaded NeoHDL was much higher than that of free [3H]IDU ( < 20% of the dose). Both parenchymal liver cells and Kupffer cells express galactose-specific receptors. By isolating liver cells after injection of the prodrug-loaded particles, it was established that hepatic uptake occurred mainly (for 84.4 +/- 3.8%) in parenchymal liver cells. Preinjection with asialofetuin substantially reduced the liver uptake of lactosylated [3H]IDU-Ol2-loaded NeoHDL, which points to uptake by the asialoglycoprotein receptor. Subcellular fractionation of the liver indicated that lactosylated [3H]IDU-Ol2-loaded NeoHDL does not merely associate to cells, but is internalized and delivered to the lysosomes. In conclusion, we show that IDU can be specifically targeted to the parenchymal liver cell. Conversion of the water-soluble parent drug into a lipophilic prodrug that is incorporated into a lactosylated reconstituted HDL particle, is an approach that may also be used to deliver other water-soluble drugs to the parenchymal liver cells. This may lead to more effective therapy for liver diseases such as hepatitis B.
- Published
- 1996
- Full Text
- View/download PDF
35. Recognition of lactoferrin and aminopeptidase M-modified lactoferrin by the liver: involvement of the remnant receptor.
- Author
-
Ziere GJ, Kruijt JK, Bijsterbosch MK, and Berkel TJ
- Subjects
- Animals, Binding Sites, Endocytosis physiology, Male, Methionyl Aminopeptidases, Rats, Rats, Wistar, Aminopeptidases blood, Lactoferrin blood, Lipoproteins, VLDL blood, Liver metabolism
- Abstract
Lactoferrin inhibits the hepatic uptake of lipoprotein remnants, and we showed earlier that arginine residues of lactoferrin are involved. In this study, lactoferrin was treated with aminopeptidase-M (APM), which resulted in removal of 14 N-terminal amino acids, including 4 clustered arginines at positions 2-5 (APM-lactoferrin). After i.v. injection into rats, 125I-APM-lactoferrin was cleared within 10 min by the liver parenchymal cells (74.7% of the dose). Binding of APM-lactoferrin to isolated parenchymal liver cells was saturable with a Kd of 186 nM (750.000 sites/cell). This is in striking contrast to the binding of lactoferrin (Kd 10 microM; 20 x 10(6) sites/cell). Preinjection of rats with 20 mg of APM-lactoferrin/kg body weight reduced the liver association of beta-VLDL by 50%, whereas lactoferrin had no effect at this dose. With isolated parenchymal liver cells, APM-lactoferrin was a more effective competitor for beta-VLDL binding than native lactoferrin (50% inhibition at 0.5 mg/ml vs. 8.0 mg/ml). We conclude that the 4-arginine cluster of lactoferrin at position 2-5 involved in the massive association of lactoferrin with the parenchymal liver cell, but is not essential for the inhibition of the lipoprotein remnant uptake. The Arg/Lys sequence at position 25-30, which resembles the binding site of apoE, may mediate the high affinity binding of lactoferrin and block the binding of beta-VLDL to the remnant receptor efficiently.
- Published
- 1996
36. Ligand specificity of the LDL-receptor related protein.
- Author
-
van Berkel TJ, Bihain BE, Ziere GJ, Kruyt JK, Bijsterbosch MK, and Kuiper J
- Subjects
- Animals, Cell Line, Humans, Low Density Lipoprotein Receptor-Related Protein-1, Species Specificity, Blood Proteins metabolism, Chylomicrons blood, Lipoproteins blood, Liver metabolism, Receptors, Immunologic metabolism
- Published
- 1996
37. Quantitative analysis of the targeting of mannose-terminal glucocerebrosidase. Predominant uptake by liver endothelial cells.
- Author
-
Bijsterbosch MK, Donker W, van de Bilt H, van Weely S, van Berkel TJ, and Aerts JM
- Subjects
- Animals, Biological Transport, Active, Endothelium cytology, Endothelium metabolism, Gaucher Disease drug therapy, Glucosylceramidase blood, Glucosylceramidase pharmacokinetics, Humans, Kupffer Cells metabolism, Liver cytology, Male, Mannose Receptor, Metabolic Clearance Rate, Rats, Rats, Wistar, Receptors, Cell Surface metabolism, Tissue Distribution, Glucosylceramidase metabolism, Lectins, C-Type, Liver metabolism, Mannose-Binding Lectins
- Abstract
Gaucher's disease is an inherited lysosomal storage disorder that is caused by a deficiency of glucocerebrosidase. The resulting accumulation of the substrate glucosylceramide in macrophages of liver, spleen, and bone marrow causes severe clinical symptoms. Gaucher's disease is treated by intravenous administration of a modified glucocerebrosidase (Alglucerase), which has exposed mannose residues to promote uptake by target macrophages. To evaluate the effectiveness of the targeting of Alglucerase, we studied the fate of the enzyme in the rat. Intravenously injected Alglucerase was rapidly cleared from the circulation (half-life 2.0 +/- 0.5 min). The liver was the main site of uptake, with 65.6 +/- 1.2% of the dose present at 10 min after injection. Smaller amounts ( < 3% of the dose) were taken up by spleen and bone marrow. Previous injection with mannan substantially increased the plasma half-life of the enzyme (14.8 +/- 3.2 min versus 1.7 +/- 0.3 min in solvent-preinjected controls) and uptake of the enzyme by liver, spleen and bone marrow was reduced by > 90%. These findings indicate that the enzyme is taken up by these organs via mannose-specific receptors. Subcellular fractionation of the liver indicated that the enzyme is internalized and transported to the lysosomes. By isolating various liver cell types after injection of the Alglucerase, it was found that endothelial cells are the main site of uptake of the enzyme: 60.8 +/- 3.4% of the total liver uptake. Parenchymal and Kupffer cells were responsible for 31.0 +/- 3.1% and 8.2 +/- 0.7% of the hepatic uptake, respectively. We conclude that Alglucerase is rapidly cleared from the circulation by mannose-specific receptors in liver, spleen, and bone marrow. However, less than 10% of the enzyme taken up by the liver is accounted for by Kupffer cells, the hepatic target cells for therapeutic intervention. It is suggested that alterations of the formulation of the therapeutic enzyme may lead to a higher uptake by Kupffer cells and other macrophages, and thus to a more (cost)effective therapy of Gaucher's disease.
- Published
- 1996
- Full Text
- View/download PDF
38. Recognition of lactoferrin and aminopeptidase M-modified lactoferrin by the liver: involvement of proteoglycans and the remnant receptor.
- Author
-
Ziere GJ, Kruijt JK, Bijsterbosch MK, and van Berkel TJ
- Subjects
- Animals, Binding Sites, Cells, Cultured, Chondroitin Lyases pharmacology, Heparin Lyase, Humans, Lipoproteins, VLDL metabolism, Male, Methionyl Aminopeptidases, Polysaccharide-Lyases pharmacology, Rats, Rats, Wistar, Receptors, Peptide metabolism, alpha-Macroglobulins metabolism, Aminopeptidases metabolism, Lactoferrin metabolism, Liver metabolism, Proteoglycans metabolism, Receptors, LDL metabolism
- Abstract
1. Lactoferrin and aminopeptidase M-modified lactoferrin (APM-lactoferrin; which lacks its 14 N-terminal amino acids) inhibit the liver uptake of lipoprotein remnant. In the present study, the role of proteoglycans in the initial interaction of beta-migrating very-low-density lipoprotein (beta-VLDL), native and APM-lactoferrin with isolated rat parenchymal liver cells was investigated. Treatment of the cells with chondroitinase lowered the Kd of lactoferrin binding (from 10 to 2.4 microM), and the number of sites/cell (from 20 x 10(6) to 7 x 10(6)), while heparinase treatment did not affect the binding. The binding characteristics of APM-lactoferrin and beta-VLDL were not altered by treatment of the cells with chondroitinase or heparinase. It is concluded that proteoglycans are not involved in the initial binding of APM-lactoferrin and beta-VLDL to parenchymal cells, while chondroitin sulphate proteoglycans are mainly responsible for the massive, low-affinity binding of native lactoferrin..2. The binding of lactoferrin, APM-lactoferrin and beta-VLDL to parenchymal liver cells was not influenced by the glutathione S-transferase-receptor-associated protein (GST-RAP) (97.2% +/- 4.0%, 95.5 +/- 3.7% and 98.5% of the control binding), while the binding of alpha 2-macroglobulin was fully blocked at 10 micrograms/ml GST-RAP (1.8 +/- 0.5% of the control binding). Since GST-RAP blocks the binding of all the known ligands to the low-density lipoprotein (LDL)-receptor-related protein (LRP), it is concluded that LRP is not the initial primary recognition site for lactoferrin, APM-lactoferrin and beta-VLDL on parenchymal liver cells. 3. We showed earlier that.APM-lactoferrin, as compared with lactoferrin, is a more effective inhibitor of the liver uptake of lipoprotein remnants (49.4 +/- 4.0% versus 80.8 +/- 4.8% of the control at 500 micrograms/ml respectively). We found in the present study that beta-VLDL is able to inhibit the binding of APM-lactoferrin to parenchymal liver cells significantly (74.9 +/- 3.3% of the control; P < 0.002), while the lactoferrin binding was unaffected. It is concluded that a still unidentified specific recognition site (the putative remnant receptor) is responsible for the initial binding of remnants to parenchymal cells and it is suggested that the partial cross-competition between APM-lactoferrin and beta-VLDL may be of further help in the elucidation of the molecular nature of this recognition site.
- Published
- 1996
- Full Text
- View/download PDF
39. Selective liver targeting of antivirals by recombinant chylomicrons--a new therapeutic approach to hepatitis B.
- Author
-
Rensen PC, van Dijk MC, Havenaar EC, Bijsterbosch MK, Kruijt JK, and van Berkel TJ
- Subjects
- Animals, Apolipoproteins E chemistry, Biological Transport, Chylomicrons pharmacokinetics, Liver metabolism, Low Density Lipoprotein Receptor-Related Protein-1, Male, Rats, Rats, Wistar, Receptors, Lipoprotein metabolism, Recombinant Proteins chemistry, Antiviral Agents administration & dosage, Chylomicrons chemistry, Drug Delivery Systems, Hepatitis B drug therapy, Idoxuridine administration & dosage
- Abstract
Hepatitis B virus (HBV) infection is the world's most important chronic virus infection. No safe and effective treatment is available at present, and clinical exploration of promising antiviral agents, such as nucleoside analogues is hampered because of significant side-effects due to their aspecific body distribution. We are exploring the possibility of the selective delivery of antiviral active drugs to liver parenchymal cells, the main site of infection and replication of HBV. Chylomicrons, which transport dietary lipids into the liver via apolipoprotein E-specific receptors, could serve as drug carriers. However, their endogenous nature hampers their application as pharmaceutical drug carriers. We report here that incorporation of a derivative of the nucleoside analogue iododeoxyuridine into recombinant chylomicrons leads to selective targeting to liver parenchymal cells. Potentially effective intracellular drug concentrations of 700 nM can be achieved, and we therefore anticipate that these drug carrier complexes represent a conceptual advance in the development of an effective and safe therapy for hepatitis B.
- Published
- 1995
- Full Text
- View/download PDF
40. Synthesis of the dioleoyl derivative of iododeoxyuridine and its incorporation into reconstituted high density lipoprotein particles.
- Author
-
Bijsterbosch MK, Schouten D, and van Berkel TJ
- Subjects
- Animals, Antiviral Agents administration & dosage, Antiviral Agents pharmacokinetics, Chemical Phenomena, Chemistry, Physical, Humans, Idoxuridine administration & dosage, Idoxuridine chemical synthesis, Idoxuridine pharmacokinetics, Injections, Intravenous, Lactose metabolism, Liver metabolism, Rats, Tritium, Antiviral Agents chemical synthesis, Drug Carriers, Idoxuridine analogs & derivatives, Idoxuridine chemistry, Lipoproteins, HDL administration & dosage, Prodrugs chemical synthesis
- Abstract
We investigated the potential use of reconstituted HDL particles (NeoHDL) as a carrier for lipophilic (pro)drugs. The antiviral drug iododeoxyuridine (IDU) was used as model compound. [3H]-IDU was derivatized with two oleoyl residues to dioleoyl[3H]iododeoxyuridine ([3H]IDU-Ol2), and the lipophilic prodrug was incorporated into NeoHDL by cosonication of [3H]IDU-Ol2 with lipids and HDL apoproteins. NeoHDL particles with the same density, size, and electrophoretic mobility as native HDL were obtained, which contained 7.3 +/- 0.8% (w/w) [3H]IDU-Ol2 (about 30 molecules of prodrug per particle). NeoHDL-associated [3H]IDU-Ol2 was stable during 2 h of incubation with human plasma; the prodrug was not appreciably hydrolyzed, nor exchanged with LDL. After intravenous injection of [3H]-IDU-Ol2-loaded 125I-NeoHDL into rats, [3H]IDU-Ol2 disappeared more rapidly from the circulation than the 125I-apoproteins (78.0 +/- 8.0% vs 30.1 +/- 4.5% of the dose cleared from plasma in 60 min, respectively). The hepatic association of the prodrug was higher than that of the apoproteins (21.6 +/- 0.5 vs 5.2 +/- 1.0% of the dose at 10 min after injection, respectively). As selective clearance and uptake of lipid esters is also observed with native HDL, this suggests that, in vivo, prodrug-loaded NeoHDL may be subject to physiological HDL-specific processing. Lactosylated [3H]IDU-Ol2-loaded 125I-NeoHDL, which contains galactose residues that can be recognized by galactose receptors on parenchymal liver cells, was rapidly cleared from plasma.(ABSTRACT TRUNCATED AT 250 WORDS)
- Published
- 1994
- Full Text
- View/download PDF
41. Specific targeting of the antiviral drug 5-iodo 2'-deoxyuridine to the parenchymal liver cell using lactosylated poly-L-lysine.
- Author
-
Biessen EA, Beuting DM, Vietsch H, Bijsterbosch MK, and Van Berkel TJ
- Subjects
- Animals, Antiviral Agents administration & dosage, Asialoglycoprotein Receptor, Asialoglycoproteins metabolism, Binding, Competitive, Deoxyuracil Nucleotides metabolism, Humans, Idoxuridine pharmacokinetics, Liver metabolism, Male, Rats, Rats, Wistar, Receptors, Cell Surface metabolism, Drug Carriers, Idoxuridine administration & dosage, Liver cytology, Polylysine chemical synthesis, Polylysine metabolism, Polylysine pharmacology
- Abstract
In this study, we describe the development and characterization of lactosylated poly-L-lysine as a potential carrier for targeting anti-viral drugs to the parenchymal liver cell. Poly-L-lysine (M(r) 38,000) was modified with 2 to 130 lactose residues per molecule poly-L-lysine. In vitro competition studies for the asialoglycoprotein receptor on parenchymal liver cells using 125I-asialoorosomucoid as radioligand revealed that mild modification of poly-L-lysine with only five lactose residues was sufficient for high affinity competition. In vivo studies showed that, after injection of poly-L-lysine modified with at least five lactose residues, about 70-80% of the injected dose was taken up by the liver. Preinjection of N-acetyl galactosamine almost completely blocked the hepatic uptake of lactosylated poly-L-lysine, indicating that galactose-recognizing receptors are involved. At 10 min following injection, the contribution of the various liver cell types to the hepatic uptake of lactosylated poly-L-lysine was determined; the parenchymal cell appeared to be responsible for more than 98% of the total liver uptake. To assess the applicability of lactosylated poly-L-lysine as an anti-viral drug carrier, it was derivatized with 4 to 15 residues of the antiviral drug 5-iodo 2'-deoxyuridine, 5'-monophosphate per molecule poly-L-lysine (4-16% by weight) via an acid-labile phosphamide bond. Maximally 0.7% of the conjugated 5-iodo 2'-deoxyuridine 5'-monophosphate was released after 1 h incubation of the drug/carrier conjugate with serum at 37 degrees C, thus establishing the stability of the conjugate in serum. The drug-carrier conjugate was rapidly cleared from the bloodstream within 1 min. Approximately 90% of the injected dose could be recovered in the liver. The parenchymal liver cell was responsible for 97% of the hepatic uptake. In vitro studies on the kinetics of endocytosis of lactosylated poly-L-lysine, derivatized with 5-iodo 2'-deoxyuridine 5'-monophosphate, by parenchymal liver cells revealed that the ligand was immediately internalized and, after a 10-min lag phase, deacetylated. Internalization and degradation did not occur in the presence of 100 mM N-acetyl galactosamine. In conclusion, the bioavailability of 5-iodo 2'-deoxyuridine 5'-monophosphate to the parenchymal liver cell is dramatically enhanced as a result of the conjugation of the anti-viral drugs to lactosylated poly-L-lysine. Accordingly, lactosylated poly-L-lysine constitutes a suitable carrier for targeting anti-viral drugs to the parenchymal liver cell.(ABSTRACT TRUNCATED AT 400 WORDS)
- Published
- 1994
- Full Text
- View/download PDF
42. Lipoprotein receptors and atherogenic receptor-mediated mechanisms.
- Author
-
van Berkel TJ, Ziere GJ, Bijsterbosch MK, and Kuiper J
- Subjects
- Animals, Cells, Cultured, Humans, Liver cytology, Liver metabolism, Protein Binding, Tumor Cells, Cultured, Arteriosclerosis metabolism, Receptors, Lipoprotein metabolism
- Abstract
The liver plays a decisive role in the regulation of the plasma levels of atherogenic lipoproteins. The primary liver interaction site of chylomicron remnants and VLDL remnants (beta-VLDL) is still unidentified, whereas the subsequent cellular uptake is likely to be mediated in concert by the LDL receptor-related protein and the LDL receptor. The nature of the primary interaction site of remnants (remnant receptor) might be a liver-specific proteoglycan or a liver-specific protein. Atherogenic modified LDL can be recognized by a family of scavenger receptors. A newly identified 95 kDa protein forms the most likely candidate for mediating the in-vivo uptake of oxidized LDL from the circulation and may, therefore, protect the body against the presence of oxidized LDL in the blood compartment.
- Published
- 1994
- Full Text
- View/download PDF
43. Removal of 14 N-terminal amino acids of lactoferrin enhances its affinity for parenchymal liver cells and potentiates the inhibition of beta- very low density lipoprotein binding.
- Author
-
Ziere GJ, Bijsterbosch MK, and van Berkel TJ
- Subjects
- Amino Acid Sequence, Aminopeptidases metabolism, Animals, Apolipoproteins E metabolism, CD13 Antigens, Cells, Cultured, Humans, Lactoferrin blood, Lipoproteins, VLDL antagonists & inhibitors, Liver cytology, Male, Molecular Sequence Data, Peptide Fragments metabolism, Protein Binding, Rats, Rats, Wistar, Sequence Deletion, Lactoferrin metabolism, Lipoproteins, VLDL metabolism, Liver metabolism
- Abstract
Lactoferrin inhibits the hepatic uptake of lipoprotein remnants, and we showed earlier that arginine residues of lactoferrin are involved. In this study, lactoferrin was treated with aminopeptidase M (APM), which resulted in removal of 14 N-terminal amino acids, including 4 clustered arginine residues at positions 2-5 (APM-lactoferrin). After intravenous injection into rats, 125I-labeled APM-lactoferrin was cleared within 10 min by the liver parenchymal cells (74.7% of the dose). In contrast to native lactoferrin, APM-lactoferrin was rapidly internalized after liver association (> 80% of the liver-associated radioactivity was internalized within 10 min). Binding of APM-lactoferrin to isolated parenchymal liver cells was saturable with a Kd of 186 nM (750,000 sites/cell). This is in striking contrast to the binding of native lactoferrin (Kd 10 microM; 20 x 10(6) sites/cell). Preinjection of rats with 20 mg of APM-lactoferrin/kg of body weight reduced the liver association of beta-very low density lipoprotein (beta-VLDL) by 50%, whereas lactoferrin had no effect at this dose. With isolated parenchymal liver cells, APM-lactoferrin was a more effective competitor for beta-VLDL binding than native lactoferrin (50% inhibition at 0.5 mg/ml versus 8.0 mg/ml). Selective modification of the arginines of APM-lactoferrin with 1,2-cyclohexanedione reduced the liver association by approximately 60% and abolished the capacity of APM-lactoferrin to inhibit the binding of 125I-labeled beta-VLDL in vitro. In conclusion, our data indicate that the four-arginine cluster of lactoferrin at positions 2-5 is involved in its massive, low affinity association of lactoferrin with the liver, possibly to proteoglycans, but is not essential for the inhibition of lipoprotein remnant uptake. The Arg-Lys sequence at positions 25-31, which resembles the binding site of apolipoprotein E, may mediate the high affinity binding of lactoferrin and block the binding of beta-VLDL to the remnant receptor with high efficiency.
- Published
- 1993
44. Development of lipoprotein-like lipid particles for drug targeting: neo-high density lipoproteins.
- Author
-
Schouten D, van der Kooij M, Muller J, Pieters MN, Bijsterbosch MK, and van Berkel TJ
- Subjects
- 3T3 Cells, Acetylgalactosamine pharmacology, Animals, Binding, Competitive, Cells, Cultured, Electrophoresis, Polyacrylamide Gel, Endothelium cytology, Endothelium metabolism, Kupffer Cells cytology, Kupffer Cells metabolism, Lipoproteins, HDL chemistry, Lipoproteins, HDL metabolism, Lipoproteins, HDL pharmacokinetics, Liver drug effects, Male, Mice, Rats, Rats, Wistar, Tissue Distribution, Drug Carriers, Lipids chemistry, Lipoproteins, HDL chemical synthesis, Liver metabolism
- Abstract
The possibility was explored of synthesizing, from commercially available lipids, high density lipoprotein (HDL)-like particles (neo-HDL) with the same physico-chemical and biological properties as native HDL. A preparation method involving egg yolk phosphatidylcholine, cholesterol, and apoproteins from HDL led to the formation of particles with a composition, size, electrophoretic mobility, and density similar to those of discoidal HDL. In vitro experiments with isolated parenchymal liver cells showed that unlabeled HDL and neo-HDL competed for the same high affinity binding sites as did radiolabeled neo-HDL, whereas an excess of unlabeled low density lipoprotein was ineffective. In vivo experiments with radio-labeled neo-HDL indicated that neo-HDL showed a slow decay upon injection into rats, whereas the liver uptake did not exceed > 10% of the injected dose. The small additional liver uptake of radioactivity from neo-HDL, compared with HDL, was due to enhanced uptake by endothelial and Kupffer cells. Lactosylation of neo-HDL led to a markedly increased decay rate and a rapid uptake by rat liver (80% in 10 min). Parenchymal cells accounted for > 90% of the total liver uptake of radiolabeled lactosylated neo-HDL. Because the liver uptake of lactosylated 125I-neo-HDL could be blocked by preinjection of N-acetylgalactosamine, we conclude that the asialoglycoprotein receptor, which is specifically localized on parenchymal liver cells, is responsible for the avid liver uptake. With a fibroblast cell line transfected with the human asialoglycoprotein receptor, it was found that lactosylated neo-HDL binds with high affinity (Kd, 40 nM), in a galactose-specific way. It can be concluded that, with commercially available lipid components, HDL-like particles (neo-HDL) with virtually the same characteristics as found for native apolipoprotein E-free HDL can be reconstituted. Lactosylated neo-HDL, which is rapidly taken up by galactose-specific receptors on parenchymal liver cells, might be used to transport antiviral drugs specifically to parenchymal liver cells.
- Published
- 1993
45. A monogalactosylated cholesterol derivative that specifically induces uptake of LDL by the liver.
- Author
-
Bijsterbosch MK, Bakkeren HF, Kempen HJ, Roelen HC, van Boom JH, and van Berkel TJ
- Subjects
- Animals, Apolipoproteins metabolism, Cells, Cultured, Cholesterol pharmacology, Liver metabolism, Male, Rats, Rats, Wistar, Receptors, Cell Surface metabolism, Cholesterol analogs & derivatives, Cholesterol Esters pharmacology, Lipoproteins, HDL metabolism, Lipoproteins, LDL metabolism, Liver drug effects, Thionucleotides pharmacology
- Abstract
We described earlier the effect of tris-gal-chol (a triantennary galactose structure coupled to cholesterol) on the fate of low density lipoprotein (LDL) and high density lipoprotein (HDL). Tris-gal-chol-loaded LDL and HDL are both efficiently cleared from blood by hepatic galactose-specific receptors. Thus, tris-gal-chol combines a beneficial LDL-reducing effect with an equally effective but undesirable HDL-lowering effect. We recently synthesized a cholesterol derivative with a single terminal galactose residue, denoted mono-gal-chol. In the present study we show that this compound, which incorporates readily into both LDL and HDL, induces rapid association of LDL and HDL to the liver. The mono-gal-chol-stimulated hepatic association of HDL, however, was about fivefold lower than that of LDL. In the liver, Kupffer cells were mainly (90%) responsible for the liver uptake of mono-gal-chol-loaded LDL, whereas the complex of mono-gal-chol with HDL was predominantly (95%) taken up by parenchymal cells. Uptake by both cell types proceeded via galactose-specific receptors and was followed by degradation of the apolipoproteins in the lysosomes. Thus, compared with tris-gal-chol, mono-gal-chol is equally effective in the induction of galactose-specific uptake of LDL by Kupffer cells. However, the galactose-specific receptor on parenchymal cells recognizes mono-gal-chol-loaded HDL less efficiently than tris-gal-chol-containing HDL. These results indicate that mono-gal-chol might be used to specifically lower LDL levels in patients with a high LDL cholesterol level.
- Published
- 1992
- Full Text
- View/download PDF
46. Lactoferrin uptake by the rat liver. Characterization of the recognition site and effect of selective modification of arginine residues.
- Author
-
Ziere GJ, van Dijk MC, Bijsterbosch MK, and van Berkel TJ
- Subjects
- Animals, Binding Sites, Biological Transport, Blood, Cyclohexanones chemistry, Humans, In Vitro Techniques, Iodine Radioisotopes, Liver cytology, Liver drug effects, Lysosomes metabolism, Male, Polysaccharides pharmacology, Rats, Rats, Inbred Strains, Tissue Distribution, Arginine metabolism, Lactoferrin metabolism, Liver metabolism
- Abstract
Recently it was found that lactoferrin, an iron-binding glycoprotein with a molecular weight of 76,500, inhibits the remnant receptor-mediated uptake of apolipoprotein E (apoE)-bearing lipoproteins by the liver. In the present study we characterized the hepatic recognition of lactoferrin. Intravenously injected 125I-lactoferrin was cleared rapidly from the circulation by the liver (92.8 +/- 9.5% of the dose at 5 min after injection). Parenchymal cells contained 97.1 +/- 1.5% of the hepatic radioactivity. Internalization, monitored by measuring the release of liver-associated radioactivity by the polysaccharide fucoidin, occurred slowly. Only about 40% of the liver-associated lactoferrin was internalized at 10 min after injection, and it took 180 min to internalize 90%. Subcellular fractionation indicated that internalized lactoferrin is transported to the lysosomes. Binding of lactoferrin to isolated parenchymal liver cells was saturable with a dissociation constant of 10 microM (20 x 10(6) binding sites/cell). The role of arginine residues on lactoferrin was studied by modifying these residues with 1,2-cyclohexanedione. The modification resulted in a strongly reduced liver association (15.9 +/- 1.6% of the dose at 5 min after injection). Furthermore, unlabeled 1,2-cyclohexanedione-modified lactoferrin did not inhibit the binding of 125I-lactoferrin to isolated parenchymal cells. Arginine residues on lactoferrin thus appear to be essential for its specific recognition by parenchymal liver cells. In particular the clustered N-terminal arginine residues, which resemble the arginine-rich receptor binding sequence in apoE, may be responsible for both the interaction of lactoferrin with its recognition site and the inhibition of the hepatic uptake of apoE-bearing lipoproteins.
- Published
- 1992
47. Lactosylated high density lipoprotein: a potential carrier for the site-specific delivery of drugs to parenchymal liver cells.
- Author
-
Bijsterbosch MK and Van Berkel TJ
- Subjects
- Animals, Cholesterol Esters pharmacokinetics, Drug Carriers metabolism, Drug Carriers pharmacokinetics, Humans, Lactose metabolism, Liver cytology, Liver ultrastructure, Male, Rats, Rats, Inbred Strains, Receptors, Cell Surface metabolism, Tissue Distribution, Tritium, Lactose pharmacokinetics, Lipoproteins, HDL metabolism, Liver metabolism
- Abstract
Mammalian liver contains two types of galactose receptors, specific for Kupffer or parenchymal cells. Because galactose-specific receptors are largely confined to the liver, galactose-bearing carriers are promising vehicles for the specific delivery of drugs to liver cells. In the present study, high density lipoprotein (HDL), a spherical particle with a diameter of 10 nm, in which a variety of lipophilic drugs can be incorporated, was provided with galactose residues by reductive lactosamination. After injection into rats, lactosylated 125I-HDL was rapidly cleared from the plasma (half-life, less than 1 min). Ten minutes after injection, the liver contained about 95% of the dose, whereas only small amounts of radioactivity were found in other tissues. The hepatic uptake was inhibited by preinjection with N-acetylgalactosamine, which indicates that the hepatic recognition sites are galactose specific. Subcellular fractionation of the liver indicated that recognition of lactosylated HDL is followed by internalization and degradation of the apoprotein in the lysosomes. Liver cells were isolated at 10 min after injection of lactosylated 125I-HDL, and it was found that uptake occurs almost exclusively by parenchymal cells. These cells contained about 98% of the hepatic radioactivity. The liver uptake of the lipid moiety of lactosylated HDL, labeled with [3H]cholesteryl oleate, was identical to that of the 125I-labeled apoproteins, which indicates that the particle is taken up as a unit. Thus, lactosylated HDL is taken up rapidly and selectively by parenchymal liver cells, and it appears that it might be a very effective vehicle for the specific delivery of lipophilic drugs to these cells.
- Published
- 1992
48. Recognition of chylomicron remnants and beta-migrating very-low-density lipoproteins by the remnant receptor of parenchymal liver cells is distinct from the liver alpha 2-macroglobulin-recognition site.
- Author
-
van Dijk MC, Ziere GJ, Boers W, Linthorst C, Bijsterbosch MK, and van Berkel TJ
- Subjects
- Animals, Cells, Cultured, Chylomicrons blood, Humans, Lactoferrin pharmacology, Liver cytology, Low Density Lipoprotein Receptor-Related Protein-1, Male, Rats, Rats, Inbred Strains, Receptors, Immunologic drug effects, Chylomicrons metabolism, Lipoproteins, VLDL metabolism, Liver metabolism, Receptors, Immunologic metabolism, alpha-Macroglobulins metabolism
- Abstract
The uptake in vivo of chylomicrons and beta-migrating very-low-density lipoprotein (beta-VLDL) by rat liver, which is primarily carried out by parenchymal cells, is inhibited, 5 min after injection, to respectively 35 and 8% of the control values after preinjection of lactoferrin. The decrease in the uptake of lipoproteins by the liver caused by lactoferrin is a specific inhibition of uptake by parenchymal cells. Competition studies in vitro demonstrate that chylomicron remnants and beta-VLDL compete for the same recognition site on parenchymal cells. Data obtained in vivo together with the competition studies performed in vitro indicate that chylomicron remnants and beta-VLDL interact specifically with the same remnant receptor. Hepatic uptake of 125I-labelled-alpha 2-macroglobulin in vivo, mediated equally by parenchymal and endothelial cells, is not decreased by preinjection of lactoferrin and no effect on the parenchymal-cell-mediated uptake is found. In vitro, alpha 2-macroglobulin and chylomicron remnants or beta-VLDL show no cross-competition. Culturing of parenchymal cells for 24-48 h leads to a decrease in the cell association of alpha 2-macroglobulin to 26% of the initial value, while the cell association of beta-VLDL with the remnant receptor is not influenced. It is concluded that beta-VLDL and chylomicron remnants are recognized by a specific remnant receptor on parenchymal liver cells, while uptake of alpha 2-macroglobulin by liver is carried out by a specific receptor system (presumably involving the LDL-receptor-related protein) which shows properties that are distinct from those of the remnant receptor.
- Published
- 1991
- Full Text
- View/download PDF
49. Enhanced hepatic uptake and processing of cholesterol esters from low density lipoprotein by specific lactosaminated Fab fragments.
- Author
-
Bijsterbosch MK, Bernini F, Bakkeren HF, Gotto AM Jr, Smith LC, and van Berkel TJ
- Subjects
- Animals, Antigen-Antibody Complex metabolism, Blood metabolism, Humans, Immunoglobulin Fab Fragments immunology, Lipoproteins, LDL immunology, Liver cytology, Rats, Rats, Inbred Strains, Tissue Distribution, Amino Sugars pharmacology, Cholesterol Esters metabolism, Immunoglobulin Fab Fragments metabolism, Lipoproteins, LDL metabolism, Liver metabolism
- Abstract
Reduction of the blood levels of low density lipoprotein (LDL) is important for lowering the incidence of atherosclerosis. In this study, LDL was directed to rat parenchymal liver cells by lactosaminated Fab fragments of anti-apolipoprotein B antibodies (LacFab). We followed the fate of intravenously injected complexes of LacFab and [3H]cholesteryl oleate-labeled LDL. Complexing of LacFab to LDL led to rapid disappearance of LDL from the circulation. At 30 minutes after injection, the liver contained 58.5 +/- 9.0% of the injected dose (at that time the liver contained only 5.7 +/- 2.2% of an injected dose of free LDL). Liver uptake was blocked by N-acetylgalactosamine but not by N-acetylglucosamine, which indicates that galactose-specific recognition sites are responsible for the LacFab-induced hepatic uptake. By isolating liver cells, it was found that parenchymal, endothelial, and Kupffer cells account for 87%, 3%, and 10% of the total hepatic uptake, respectively. Subcellular fractionation of the liver indicated that the complexes are rapidly internalized and transported to lysosomes. Within 1 hour after injection, virtually all the [3H]cholesteryl oleate of the internalized LDL was hydrolyzed; hydrolysis was followed by excretion of radioactivity into the bile. Compared with rats injected with native [3H]cholesteryl oleate-labeled LDL, eight times as much radioactivity was excreted into the bile during the first 4 hours after the injection of LacFab-complexed [3H]cholesteryl oleate-labeled LDL. Thus, LacFab induces enhanced hepatic uptake of LDL via galactose receptors on the parenchymal cells, followed by processing in lysosomes and excretion into the bile. In this way, LacFab induces an increased irreversible removal of LDL cholesterol from the body.
- Published
- 1991
- Full Text
- View/download PDF
50. Water-soluble cholesteryl-containing phosphorothioate monogalactosides: synthesis, properties, and use in lowering blood cholesterol by directing plasma lipoproteins to the liver.
- Author
-
Roelen HC, Bijsterbosch MK, Bakkeren HF, van Berkel TJ, Kempen HJ, Buytenhek M, van der Marel GA, and van Boom JH
- Subjects
- Chemical Phenomena, Chemistry, Cholesterol blood, Cholesterol chemical synthesis, Cholesterol pharmacology, Galactosides blood, Galactosides pharmacology, Humans, Lipoproteins metabolism, Lipoproteins, HDL blood, Lipoproteins, HDL metabolism, Lipoproteins, LDL blood, Lipoproteins, LDL metabolism, Molecular Structure, Organothiophosphorus Compounds blood, Organothiophosphorus Compounds pharmacology, Solubility, Water, Anticholesteremic Agents chemical synthesis, Cholesterol analogs & derivatives, Galactosides chemical synthesis, Lipoproteins blood, Liver metabolism, Organothiophosphorus Compounds chemical synthesis
- Abstract
The synthesis of several monogalactoside-terminated phosphorothiolated cholesteryl derivatives is described. Monogalactosyl derivatives are coupled by phosphorothiolation to cholesterol by using ethylene glycol units as hydrophilic spacer moieties. The resulting compounds are easily soluble in water. Upon addition of such solutions to human serum (to 2 mM final concentration) the compounds are readily incorporated into lipoproteins. Isolated low-density lipoprotein (LDL) and high-density lipoprotein (HDL), preloaded with the compounds, are rapidly cleared from the circulation by the liver. The hepatic association is blocked by N-acetylgalactosamine, which indicates that galactose-specific recognition sites are responsible for the increased liver uptake. The plasma clearance and hepatic uptake of LDL loaded with the compounds is substantially higher (about 2-fold) than clearance and uptake of HDL containing the compounds. The selectivity of the effects of monogalactoside-terminated phosphorothiolated cholesteryl derivatives on the in vivo behavior of LDL as compared to that of HDL indicates that these compounds might be used to lower specifically LDL levels in patients with a high LDL-cholesterol level.
- Published
- 1991
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.