Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Nucleic Acid Ther ; 27(3): 130-143, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28118087

ABSTRACT

Splice-switching antisense oligonucleotides are emerging treatments for neuromuscular diseases, with several splice-switching oligonucleotides (SSOs) currently undergoing clinical trials such as for Duchenne muscular dystrophy (DMD) and spinal muscular atrophy (SMA). However, the development of systemically delivered antisense therapeutics has been hampered by poor tissue penetration and cellular uptake, including crossing of the blood-brain barrier (BBB) to reach targets in the central nervous system (CNS). For SMA application, we have investigated the ability of various BBB-crossing peptides for CNS delivery of a splice-switching phosphorodiamidate morpholino oligonucleotide (PMO) targeting survival motor neuron 2 (SMN2) exon 7 inclusion. We identified a branched derivative of the well-known ApoE (141-150) peptide, which as a PMO conjugate was capable of exon inclusion in the CNS following systemic administration, leading to an increase in the level of full-length SMN2 transcript. Treatment of newborn SMA mice with this peptide-PMO (P-PMO) conjugate resulted in a significant increase in the average lifespan and gains in weight, muscle strength, and righting reflexes. Systemic treatment of adult SMA mice with this newly identified P-PMO also resulted in small but significant increases in the levels of SMN2 pre-messenger RNA (mRNA) exon inclusion in the CNS and peripheral tissues. This work provides proof of principle for the ability to select new peptide paradigms to enhance CNS delivery and activity of a PMO SSO through use of a peptide-based delivery platform for the treatment of SMA potentially extending to other neuromuscular and neurodegenerative diseases.


Subject(s)
Apolipoproteins E/pharmacokinetics , Morpholinos/pharmacology , Morpholinos/pharmacokinetics , Muscular Atrophy, Spinal/drug therapy , Peptides/pharmacokinetics , Animals , Animals, Newborn , Apolipoproteins E/chemical synthesis , Apolipoproteins E/chemistry , Biomarkers/blood , Blood-Brain Barrier/chemistry , Blood-Brain Barrier/metabolism , Brain/cytology , Cell Survival/drug effects , Disease Models, Animal , Exons , Fibroblasts/drug effects , Hepatocytes/drug effects , Humans , Kidney/chemistry , Mice , Morpholinos/chemistry , Morpholinos/therapeutic use , Nanoconjugates/analysis , Nanoconjugates/chemistry , Nanoconjugates/therapeutic use , Peptides/chemical synthesis , Peptides/chemistry , Phenotype , Quadriceps Muscle/chemistry , Survival of Motor Neuron 2 Protein/drug effects
2.
FEBS J ; 283(11): 2115-31, 2016 06.
Article in English | MEDLINE | ID: mdl-27028511

ABSTRACT

Cationic antimicrobial peptides (AMPs) possess fast and broad-spectrum activity against both Gram-negative and Gram-positive bacteria, as well as fungi. It has become increasingly evident that many AMPs, including those that derive from fragments of host proteins, are multifunctional and able to mediate various immunomodulatory functions and angiogenesis. Among these, synthetic apolipoprotein-derived peptides are safe and well tolerated in humans and have emerged as promising candidates in the treatment of various inflammatory conditions. Here, we report the characterization of a new AMP corresponding to residues 133-150 of human apolipoprotein E. Our results show that this peptide, produced either by chemical synthesis or by recombinant techniques in Escherichia coli, possesses a broad-spectrum antibacterial activity. As shown for several other AMPs, ApoE (133-150) is structured in the presence of TFE and of membrane-mimicking agents, like SDS, or bacterial surface lipopolysaccharide (LPS), and an anionic polysaccharide, alginate, which mimics anionic capsular exo-polysaccharides of several pathogenic microorganisms. Noteworthy, ApoE (133-150) is not toxic toward several human cell lines and triggers a significant innate immune response, assessed either as decreased expression levels of proinflammatory cytokines in differentiated THP-1 monocytic cells or by the induction of chemokines released from PBMCs. This novel bioactive AMP also showed a significant anti-inflammatory effect on human keratinocytes, suggesting its potential use as a model for designing new immunomodulatory therapeutics.


Subject(s)
Antimicrobial Cationic Peptides/genetics , Apolipoproteins E/genetics , Immunity, Innate/drug effects , Immunologic Factors/genetics , Amino Acid Sequence/genetics , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/chemical synthesis , Antimicrobial Cationic Peptides/administration & dosage , Antimicrobial Cationic Peptides/chemical synthesis , Apolipoproteins E/administration & dosage , Apolipoproteins E/chemical synthesis , Apolipoproteins E/chemistry , Escherichia coli/genetics , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Humans , Immunologic Factors/administration & dosage , Immunologic Factors/chemical synthesis , Lipopolysaccharides/genetics , Lipopolysaccharides/metabolism , Microbial Sensitivity Tests
3.
Hepatology ; 56(2): 484-91, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22334503

ABSTRACT

UNLABELLED: Hepatitis C virus (HCV) entry is a multiple-step process involving a number of host factors and hence represents a promising target for new antiviral drug development. In search of novel inhibitors of HCV infection, we found that a human apolipoprotein E (apoE) peptide, hEP, containing both a receptor binding fragment and a lipid binding fragment of apoE specifically blocked the entry of cell culture grown HCV (HCVcc) at submicromolar concentrations. hEP caused little cytotoxicity in vitro and remained active even if left 24 hours in cell culture. Interestingly, hEP inhibited neither human immunodeficiency virus (HIV)-HCV pseudotypes (HCVpp) nor HIV and Dengue virus (DENV) infection. Further characterization mapped the anti-HCV activity to a 32-residue region that harbors the receptor binding domain of apoE, but this fragment must contain a cysteine residue at the N-terminus to mediate dimer formation. The anti-HCV activity of the peptide appears to be dependent on both its length and sequence and correlates with its ability to bind lipids. Finally, we demonstrated that the apoE-derived peptides directly blocked the binding of both HCVcc and patient serum-derived virus to hepatoma cells as well as primary human hepatocytes. CONCLUSION: apoE peptides potently inhibit HCV infection and suggest a direct role of apoE in mediating HCV entry. Our findings also highlight the potential of developing apoE mimetic peptides as novel HCV entry inhibitors by targeting HCV-host interactions.


Subject(s)
Apolipoproteins E/pharmacology , Hepacivirus/drug effects , Hepatitis C/drug therapy , Hepatocytes/virology , Peptides/pharmacology , Virus Internalization/drug effects , Amino Acid Sequence , Antiviral Agents/pharmacology , Apolipoproteins E/chemical synthesis , Cholesterol/metabolism , HEK293 Cells , Hepacivirus/growth & development , Hepacivirus/metabolism , Hepatitis C/virology , Hepatocytes/cytology , Hepatocytes/metabolism , Host-Pathogen Interactions/drug effects , Humans , Liposomes/metabolism , Molecular Sequence Data , Peptides/chemical synthesis , Receptors, LDL/metabolism
4.
Behav Brain Res ; 214(2): 395-401, 2010 Dec 25.
Article in English | MEDLINE | ID: mdl-20600347

ABSTRACT

COG1410, a small, novel ApoE-mimetic peptide derived from the receptor binding region of apolipoprotein E (ApoE), has been classified as anti-inflammatory in nature and improves motor, sensorimotor, and cognitive dysfunction following cortical contusion injury (CCI). In order to further examine COG1410's preclinical efficacy on cognitive recovery, the present study evaluated COG1410 following moderate fluid percussion injury (FPI). Animals were prepared with a moderate, unilateral FPI over the hippocampus. Following FPI, animals received a regimen of five doses of COG1410 or vehicle at 2 and 4h (1.0mg/kg, i.v.) followed by additional doses administered 24, 48, and 72 h (1.0mg/kg, i.p.). Prior to injury, animals were trained for 4 days (4 trials/day) in the Morris water maze (MWM) and then tested for retrograde amnesia on post-FPI day 11 and then on a working memory task on day 18. Testing for motor dysfunction on the tapered balanced beam began on day 2 post-FPI. Administration of this regimen of COG1410 significantly improved retention of memory in the retrograde amnesia test compared to vehicle post-FPI. However, COG1410 did not significantly improve acquisition of working memory in the MWM. Motor dysfunction on the tapered beam post-FPI was improved in the COG1410-treated group compared to vehicle treatment. Cortical lesion analysis revealed that the COG1410-treated animals demonstrated significantly less tissue loss compared to vehicle-treated animals. The results of this study suggest that COG1410 significantly limited the behavioral dysfunction and tissue loss associated with FPI and demonstrated continued preclinical efficacy for TBI.


Subject(s)
Apolipoproteins E/administration & dosage , Brain Injuries/drug therapy , Cerebral Cortex/pathology , Cognition Disorders/drug therapy , Memory Disorders/drug therapy , Recovery of Function/drug effects , Animals , Apolipoproteins E/chemical synthesis , Brain Injuries/complications , Brain Injuries/pathology , Cognition Disorders/complications , Cognition Disorders/pathology , Disease Models, Animal , Drug Administration Schedule , Hippocampus/injuries , Injections, Intraperitoneal , Injections, Intravenous , Male , Memory Disorders/complications , Rats , Rats, Long-Evans
5.
J Neurotrauma ; 24(7): 1093-107, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17610350

ABSTRACT

Traumatic brain injury (TBI) is a silent epidemic affecting approximately 1.4 million Americans annually, at an estimated annual cost of $60 billion in the United States alone. Despite an increased understanding of the pathophysiology of closed head injury, there remains no pharmacological intervention proven to improve functional outcomes in this setting. Currently, the existing standard of care for TBI consists primarily of supportive measures. Apolipoprotein E (apoE) is the primary apolipoprotein synthesized in the brain in response to injury, where it modulates several components of the neuroinflammatory cascade associated with TBI. We have previously demonstrated that COG133, an apoE mimetic peptide, improved functional outcomes and attenuated neuronal death when administered as a single intravenous injection at 30 min post-TBI in mice. Using the principles of rational drug design, we developed a more potent analog, COG1410, which expands the therapeutic window for the treatment of TBI by a factor of four, from 30 min to 2 h. Mice that received a single intravenous injection of COG1410 at 120 min post-TBI exhibited significant improvement on a short term test of vestibulomotor function and on a long term test of spatial learning and memory. This was associated with a significant attenuation of microglial activation and neuronal death in the hippocampus, the neuroanatomical substrate for learning and memory. Rationally derived apoE mimetic peptides have been demonstrated to exert neuroprotective and anti-inflammatory effects in vitro and in clinically relevant models of brain injury. This represents a novel therapeutic strategy in the treatment of TBI.


Subject(s)
Apolipoproteins E/antagonists & inhibitors , Brain Injuries/drug therapy , Brain/drug effects , Encephalitis/drug therapy , Peptides/pharmacology , Recovery of Function/drug effects , Animals , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Apolipoproteins E/chemical synthesis , Apolipoproteins E/metabolism , Apolipoproteins E/pharmacology , Apolipoproteins E/therapeutic use , Brain/metabolism , Brain/physiopathology , Brain Injuries/metabolism , Brain Injuries/physiopathology , Cell Line , Disease Models, Animal , Encephalitis/metabolism , Encephalitis/physiopathology , Gliosis/drug therapy , Gliosis/etiology , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/physiopathology , Injections, Intravenous , Male , Memory Disorders/drug therapy , Memory Disorders/etiology , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/pathology , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/pharmacology , Peptides/chemical synthesis , Peptides/therapeutic use , Recovery of Function/physiology , Time Factors , Treatment Outcome
6.
Atherosclerosis ; 194(1): 116-24, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17126342

ABSTRACT

OBJECTIVE: Apolipoprotein E (ApoE) is a lipid transport protein with expanded functions in cellular responses to tissue injury, immune regulation and cell growth. ApoE directs vascular changes that contribute to arterial protection as evidenced by the fact that isoforms of ApoE and ApoE deficiency correlate closely with accelerated plaque growth. The N-terminus of the ApoE protein has well-characterized functions, displaying lipid-binding and anti-atherogenic activity, whereas the function of the C-terminus is only partially defined. We have assessed the effects of a 14 amino acid C-terminal ApoE peptide, termed Ep1.B (239-252), on intimal neoplasia in animal models. This peptide is a fragment of a naturally processed peptide (236-252) of murine ApoE. METHODS AND RESULTS: Ep1.B injection reduced neointimal hyperplasia after vascular surgery in rats and mice. When given during early plaque progression in ApoE-deficient mice, Ep1.B injections also prevented plaque growth. Treatment with Ep1.B did not, however, reduce established plaque growth in older mice. Peptides with alanine substitution of amino acid 249, Ep1.N, and with complete sequence reversal, Ep1.R, did not consistently inhibit plaque growth. CONCLUSION: A naturally processed C-terminal ApoE peptide, Ep1.B, has anti-atherogenic activity indicating a role for this naturally metabolized peptide in vascular wound healing and lipid homeostasis.


Subject(s)
Apolipoproteins E/pharmacology , Atherosclerosis/drug therapy , Atherosclerosis/pathology , Peptide Fragments/pharmacology , Angioplasty/adverse effects , Animals , Apolipoproteins E/chemical synthesis , Apolipoproteins E/chemistry , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Atherosclerosis/immunology , Carotid Artery Injuries/immunology , Carotid Artery Injuries/pathology , Disease Models, Animal , Female , Femoral Artery/immunology , Femoral Artery/injuries , Femoral Artery/pathology , Histocompatibility Antigens Class II/metabolism , Hyperplasia , Iliac Artery/injuries , Iliac Artery/pathology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Peptide Fragments/chemical synthesis , Peptide Fragments/metabolism , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , T-Lymphocytes/pathology , Tunica Intima/pathology
7.
Biochemistry ; 43(23): 7328-35, 2004 Jun 15.
Article in English | MEDLINE | ID: mdl-15182176

ABSTRACT

LRP1 is a cell surface receptor responsible for clearing some 30 known ligands. We have previously shown that each of the three complete LDL receptor-homology domains of the LRP1 extracellular domain (sLRPs) binds apoE-enriched beta-VLDL particles. Here we show that two peptides from the N-terminal receptor binding domain of apoE, which are known to elicit a number of different cellular responses, bind to LRP1. Solution binding assays show that the two peptides, apoE(130-149) and apoE(141-155)(2), interact with each of the sLRPs (2, 3, and 4). Each peptide was found to exhibit the same solution binding characteristics as apoE-enriched beta-VLDL particles. Surface plasmon resonance analyses of the sLRP-apoE peptide interaction show that both peptides bind the sLRPs with K(D) values in the 100 nM range, a value similar to the effective concentration required for observation of the cellular responses. Consistent with results from mutagenesis studies of binding of apoE to LDLR, apoE(130-149,Arg142Glu) bound with a K(D) similar to that of the wild-type sequence, while apoE(130-149,Lys143Glu) showed a 10-fold decrease in K(D). Each of the peptides bound heparin, and heparin competed for sLRP binding.


Subject(s)
Apolipoproteins E/chemistry , Apolipoproteins E/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Molecular Mimicry , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Amino Acid Sequence , Apolipoproteins E/chemical synthesis , Apolipoproteins E/genetics , Binding, Competitive , Cell Line , Circular Dichroism , Heparin/metabolism , Humans , Kinetics , Lipoproteins, VLDL/metabolism , Liposomes/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/chemistry , Molecular Sequence Data , Peptide Fragments/chemical synthesis , Peptide Fragments/genetics , Precipitin Tests , Protein Binding , Solutions , Surface Plasmon Resonance
8.
Biochemistry ; 40(46): 14020-9, 2001 Nov 20.
Article in English | MEDLINE | ID: mdl-11705394

ABSTRACT

The pi-helix is a secondary structure with 4.4 amino acids per helical turn. Although it was proposed in 1952, no experimental support for its existence was obtained until the mid-1980s. While short peptides are unlikely to assume a marginally stable secondary structure spontaneously, they might do so in the presence of appropriate structural constraints. In this paper, we describe a peptide that is designed to assume a pi-helical conformation when stabilized by cetyltrimethylammonium bromide (CTAB) micelles and Zn(2+). In the designed peptide, lipophilic amino acids are placed such that it would be amphiphilic in the pi-helical, but not in the alpha-helical, conformation. Also, two His residues are incorporated with i, i + 5 spacing, designed to allow binding of Zn(2+) in a pi-helical but not an alpha-helical conformation. The peptide was found to form moderately stable monolayers at the air-water interface, with a collapse pressure that almost doubled when there was Zn(2+) in the subphase. Also, CTAB micelles induced a marked increase in the helicity of the peptide. In 50% TFE, the peptide had a CD spectrum consistent with an alpha-helical structure. The addition of 1 mM Zn(2+) to this solvent caused a saturable decline in ellipticity to approximately half of its original value. The peptide also bound Zn(2+) when it was bound to CTAB micelles, with Zn(2+) again inducing a decrease in ellipticity. The peptide had slightly greater affinity for Zn(2+) in the presence of the CTAB than in a 50% TFE solution (K(d) = 3.1 x 10(-4) M in CTAB and 2.3 x 10(-4) M in TFE). van't Hoff analysis indicated that thermal denaturation of the peptide in 50% TFE containing 1 mM Zn(2+) was associated with both enthalpic and entropic changes that were greater than those in the absence of Zn(2+). These observations are all consistent with the proposal that the peptide assumed a pi-helical conformation in the presence of Zn(2+) and CTAB micelles, and has allowed the stability of this rare conformation to be assessed.


Subject(s)
Peptides/chemical synthesis , Peptides/metabolism , Zinc/metabolism , Amino Acid Sequence , Amino Acid Substitution , Apolipoproteins E/chemical synthesis , Apolipoproteins E/metabolism , Cetrimonium , Cetrimonium Compounds/pharmacology , Circular Dichroism , Enzyme Stability , Humans , Molecular Sequence Data , Pressure , Protein Binding/drug effects , Protein Engineering , Protein Structure, Secondary/drug effects , Surface Properties , Thermodynamics
9.
J Clin Invest ; 101(1): 223-34, 1998 Jan 01.
Article in English | MEDLINE | ID: mdl-9421485

ABSTRACT

Apolipoprotein (apo) E mediates lipoprotein binding to cellular lipoprotein receptors. Previously we reported that a synthetic peptide representing a linear dimeric repeat of amino acids 141-155 binds cellular LDL receptors. To prepare an apoE peptide that bound to both cholesterol-rich lipoproteins and lipoprotein receptors, an NH2-terminal acetylated apoE dimer peptide was synthesized. This acetylated peptide preferentially associated with lipoproteins in plasma, whereas nonacylated peptides were poor lipid binders. Acetylated peptide/LDL complexes (molar ratios of 4-5:1) enhanced the interaction of LDL with cultured human fibroblasts by 7-12-fold. Participation by both receptors and cell surface heparin sulfate proteoglycans was observed. When a preformed peptide/125I-LDL complex was injected intravenously into C57BL/6J apoE-deficient mice, its rate of removal was threefold higher than that of 125I-LDL alone. The liver and the spleen were major tissue distribution sites. Intravenous administration of free acetylated peptide resulted in a 30% reduction in total plasma cholesterol within 3-30 min, which reflected a 40-50% and 20-26% reduction in very low density lipoproteins and intermediate density lipoproteins, respectively. Therefore, this peptide selectively associated with cholesterol-rich lipoproteins and mediated their acute clearance in vivo.


Subject(s)
Apolipoproteins E/metabolism , Cholesterol/metabolism , Lipoproteins/metabolism , Peptides/metabolism , Animals , Apolipoproteins E/chemical synthesis , Apolipoproteins E/pharmacology , Chromatography, Agarose , Electrophoresis, Agar Gel , Humans , Lipoproteins, LDL/metabolism , Mice , Mice, Inbred C57BL , Peptides/chemical synthesis , Peptides/pharmacology , Tissue Distribution
10.
Mol Pharmacol ; 52(3): 445-55, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9281607

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.


Subject(s)
Antineoplastic Agents/administration & dosage , Apolipoproteins E/chemical synthesis , Apolipoproteins E/metabolism , Lipoproteins, LDL/administration & dosage , Liposomes/chemical synthesis , Liposomes/metabolism , 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
11.
J Biol Chem ; 266(34): 22803-6, 1991 Dec 05.
Article in English | MEDLINE | ID: mdl-1744074

ABSTRACT

Human plasma apolipoprotein E (apoE) is a low density lipoprotein (LDL) receptor ligand. It targets cholesterol-rich lipoproteins to LDL receptors on both hepatic and peripheral cells. The region of apoE responsible for its binding to the LDL receptor has been localized to amino acids 140-160. An apoE 141-155 monomeric peptide and a dimeric 141-155 tandem peptide were synthesized and tested for their inhibition of 125I-LDL degradation by human fibroblasts and human monocytic-like cells, THP-1. The monomer had no activity at 250 microM, but the dimer inhibited 125I-LDL degradation by 50% at 5 microM. The inhibition was specific for the LDL receptor because the dimer did not inhibit the degradation of 125I-acetylated LDL by scavenger receptors expressed by phorbol ester-stimulated THP-1 cells. As reported for native apoE, amino acid substitutions of Lys-143----Ala, Leu-144----Pro, and Arg-150----Ala decreased the inhibitory effectiveness of the dimer. Furthermore, a trimer of the 141-155 sequence had a 20-fold greater inhibitory activity than the dimer. Studies with a radioiodinated dimer indicated that some of the inhibitory activity could be a result of the interaction of the dimer with LDL. However, direct binding of the 125I-dimeric peptide to THP-1 cells was observed as well. This binding was time-dependent, linear with increasing cell number, Ca(2+)- but not Mg(2+)-dependent, saturable, inhibited by lipoproteins, and increased by preculture of the cells in lipoprotein-depleted medium. Therefore, a synthetically prepared dimeric repeat of amino acid residues 141-155 of apoE binds the LDL receptor.


Subject(s)
Apolipoproteins E/chemical synthesis , Receptors, LDL/metabolism , Amino Acid Sequence , Apolipoproteins E/metabolism , Binding, Competitive , Cell Line , Fibroblasts/metabolism , Humans , Molecular Sequence Data , Monocytes/metabolism , Peptides/chemical synthesis
12.
J Biol Chem ; 265(1): 490-2, 1990 Jan 05.
Article in English | MEDLINE | ID: mdl-2294117

ABSTRACT

Apolipoprotein E is a plasma protein comprised of a lipid binding region (which together with other apoproteins maintains the structure of lipoprotein particles) and a receptor binding domain (which interacts with cellular receptors for control of triglyceride and cholesterol metabolism). A peptide, comprising residues 129-169 of human apolipoprotein E, which contains both a putative lipid-binding region and receptor binding domain, has been synthesized by solid phase techniques. Diffraction quality crystals of the synthetic apolipoprotein E fragment129-169 have been obtained at room temperature by vapor diffusion with polyethylene glycol in the presence of the nonionic detergent beta-octylglucoside. The crystals have been characterized with x-radiation as orthorhombic, space group I222 or I2(1)2(1)2(1), with unit cell dimensions a = 61.91, b = 30.84, and c = 42.79 A. There are eight molecules per unit cell, with one molecule (Mr = 4771) in each asymmetric unit. Precession photographs show that crystals diffract beyond 2.7-A resolution and are stable in the x-ray beam at room temperature for at least 200 h; thus, they can be used to collect three-dimensional data for a detailed crystallographic analysis.


Subject(s)
Apolipoproteins E/chemical synthesis , Peptide Fragments/chemical synthesis , X-Ray Diffraction , Crystallization , Glucosides , Humans , Polyethylene Glycols
SELECTION OF CITATIONS
SEARCH DETAIL
...