Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-32462055

ABSTRACT

A growing body of literature supports the role of apolipoproteins present in HDL in the treatment of pro-inflammatory diseases including cancer. We examined whether bovine HDL (bHDL) and three dual-domain peptides, namely AEM-28 and its analog AEM-28-2, and HM-10/10, affect tumor growth and development in mouse models of ovarian and colon cancer. We demonstrate that bHDL inhibits mouse colorectal cancer cell line CT26-mediated lung tumor development, and mouse ovarian cancer cell line ID8-mediated tumor burden. We also demonstrate that, although to different degrees, dual-domain peptides inhibit cell viability of mouse and human ovarian and colon cancer cell lines, but not that of normal human colonic epithelial cells or NIH3T3 mouse fibroblasts. Dual-domain peptides administered subcutaneously or in a chow diet decrease CT26 cell-mediated tumor burden, tumor growth, and tumor dissemination in BALB/c mice. Plasma levels of lysophosphatidic acid (LPA) are significantly reduced in mice that received bHDL and the dual-domain peptides, suggesting that reduction by effecting accumulation and/or synthesis of pro-inflammatory lipids may be one of the mechanisms for the inhibition of tumor development by bHDL and the dual-domain peptides. Our studies suggest that therapeutics based on apolipoproteins present in HDL may be novel agents for the treatment of epithelial adenocarcinomas of the ovary and colon.

2.
J Clin Invest ; 129(9): 3670-3685, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31184596

ABSTRACT

Cyclooxygenase 2 (Cox2) total knockout and myeloid knockout (MKO) mice develop Crohn's-like intestinal inflammation when fed cholate-containing high fat diet (CCHF). We demonstrated that CCHF impaired intestinal barrier function and increased translocation of endotoxin, initiating TLR/MyD88-dependent inflammation in Cox2 KO but not WT mice. Cox2 MKO increased pro-inflammatory mediators in LPS-activated macrophages, and in the intestinal tissue and plasma upon CCHF challenge. Cox2 MKO also reduced inflammation resolving lipoxin A4 (LXA4) in intestinal tissue, while administration of an LXA4 analog rescued disease in Cox2 MKO mice fed CCHF. The apolipoprotein A-I (APOA1) mimetic 4F mitigated disease in both the Cox2 MKO/CCHF and piroxicam-accelerated Il10-/- models of inflammatory bowel disease (IBD) and reduced elevated levels of pro-inflammatory mediators in tissue and plasma. APOA1 mimetic Tg6F therapy was also effective in reducing intestinal inflammation in the Cox2 MKO/CCHF model. We further demonstrated that APOA1 mimetic peptides: i) inhibited LPS and oxidized 1-palmitoyl-2-arachidonoyl-sn-phosphatidylcholine (oxPAPC) dependent pro-inflammatory responses in human macrophages and intestinal epithelium; and ii) directly cleared pro-inflammatory lipids from mouse intestinal tissue and plasma. Our results support a causal role for pro-inflammatory and inflammation resolving lipids in IBD pathology and a translational potential for APOA1 mimetic peptides for the treatment of IBD.


Subject(s)
Apolipoprotein A-I/pharmacology , Cyclooxygenase 2/genetics , Inflammatory Bowel Diseases/drug therapy , Intestines/pathology , Animals , Disease Models, Animal , Endotoxins/metabolism , Female , Humans , Inflammatory Bowel Diseases/pathology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxygen/metabolism , Peptides/chemistry , Permeability , Piroxicam/pharmacology , Receptors, Formyl Peptide/metabolism , Signal Transduction
3.
Chem Phys Lipids ; 219: 28-35, 2019 03.
Article in English | MEDLINE | ID: mdl-30707910

ABSTRACT

High density lipoprotein (HDL) is prone to modification by the oxidizing and chlorinating agent hypochlorite anion (OCl-). Oxidation of apolipoprotein (apo) A-I, the major protein in HDL, reduces ABCA-1 mediated cholesterol efflux and other protective responses to HDL. The apoA-I mimetic peptide 4F has been shown to undergo oxidation; however, the ability of the peptide to mediate cholesterol efflux remains intact. Here, we show that 4F protects apoA-I from hypochlorite-mediated oxidation. Mass spectral analysis of apoA-I shows that tyrosine residues that are prone to hypochlorite-mediated chlorination are protected in the presence of 4F. Furthermore, 4F enhances the cholesterol efflux ability of apoA-I to a greater extent than either 4F or apoA-I alone, even after hypochlorite oxidation. These observations suggest that apoA-I in lipid complexes may be protected by the presence of 4F, resulting in the preservation of its anti-inflammatory and anti-atherogenic properties. These studies also form the basis for the future studies of nanoparticles possessing both apoA-I and 4F.


Subject(s)
Apolipoprotein A-I/chemistry , Peptides/chemistry , ATP Binding Cassette Transporter 1/metabolism , Amino Acid Sequence , Apolipoprotein A-I/analysis , Cell Line , Cholesterol/metabolism , Humans , Hypochlorous Acid/chemistry , Mass Spectrometry , Oxidation-Reduction , Phosphatidylcholines/chemistry
4.
J Lipid Res ; 59(11): 2075-2083, 2018 11.
Article in English | MEDLINE | ID: mdl-30201630

ABSTRACT

Ac-hE18A-NH2 is a dual-domain apoE mimetic peptide that possesses the putative receptor binding domain from apoE (LRKLRKRLLR, denoted hE; residues 141-150) covalently attached to lipid-associating peptide 18A. Like apoE, Ac-hE18A-NH2 reduces plasma cholesterol in animal models and exhibits anti-inflammatory properties independent of its cholesterol-reducing effect. Ac-hE18A-NH2 has already undergone phase I clinical trials as a lipid-lowering agent. To explore the therapeutic potential more, we designed and synthesized new analogues by linking ɑ-aminohexanoic acid, octanoic acid, or myristic acid to LRRLRRRLLR-18A-NH2 ([R]hE18A-NH2) and examined the cholesterol-lowering potency in animals. The modified peptides effectively reduced plasma cholesterol in apoE-null mice fed standard chow or a Western diet; the myristyl analogue was the most effective. A single administration of the myristyl analogue reduced plasma total and LDL cholesterol in a dose-dependent manner in hypercholesterolemic cynomolgus macaques for up to 1 week despite the continuation of a cholesterol-supplemented diet. The myristyl peptide (7.4 mg/kg) reduced total and LDL cholesterol at 24 h by 64% and 74%, respectively; plasma HDL levels were modestly reduced and returned to baseline by day 7. These new analogues should exhibit enhanced potency at lower doses than Ac-hE18A-NH2, which may make them attractive therapeutic candidates for clinical trials.


Subject(s)
Apolipoproteins E/chemistry , Cholesterol/blood , Peptides/chemistry , Peptides/pharmacology , Animals , Cholesterol, LDL/blood , Chromatography, High Pressure Liquid , Electrophoresis, Agar Gel , Female , Haplorhini , Humans , Intercellular Signaling Peptides and Proteins , Lipid Metabolism/drug effects , Macaca , Male , Mice , Mice, Knockout , Peptides/blood
5.
Structure ; 23(7): 1214-26, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26095027

ABSTRACT

Biogenesis of high-density lipoproteins (HDL) is coupled to the transmembrane protein, ATP-binding cassette transporter A1 (ABCA1), which transports phospholipid (PL) from the inner to the outer membrane monolayer. Using a combination of computational and experimental approaches, we show that increased outer lipid monolayer surface density, driven by excess PL or membrane insertion of amphipathic helices, results in pleating of the outer monolayer to form membrane-attached discoidal bilayers. Apolipoprotein (apo)A-I accelerates and stabilizes the pleats. In the absence of apoA-I, pleats collapse to form vesicles. These results mimic cells overexpressing ABCA1 that, in the absence of apoA-I, form and release vesicles. We conclude that the basic driving force for nascent discoidal HDL assembly is a PL pump-induced surface density increase that produces lipid monolayer pleating. We then argue that ABCA1 forms an extracellular reservoir containing an isolated pressurized lipid monolayer decoupled from the transbilayer density buffering of cholesterol.


Subject(s)
Lipid Bilayers/chemistry , Lipoproteins, HDL/chemistry , Phosphatidylcholines/chemistry , ATP Binding Cassette Transporter 1/chemistry , Cell Membrane Structures/chemistry , Cholesterol/chemistry , Molecular Dynamics Simulation
6.
Inflammation ; 37(5): 1401-12, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24647607

ABSTRACT

Human apolipoprotein A-I (apoA-I) mimetic L-4F inhibits acute inflammation in endotoxemic animals. Since neutrophils play a crucial role in septic inflammation, we examined the effects of L-4F, compared to apoA-I, on lipopolysaccharide (LPS)-mediated activation of human neutrophils. We performed bioassays in human blood, isolated human neutrophils (incubated in 50 % donor plasma), and isolated human leukocytes (incubated in 5 and 50 % plasma) in vitro. In whole blood, both L-4F and apoA-I inhibited LPS-mediated elevation of TNF-α and IL-6. In LPS-stimulated neutrophils, L-4F and apoA-I (40 µg/ml) also decreased myeloperoxidase and TNF-α levels; however, L-4F tended to be superior in inhibiting LPS-mediated increase in IL-6 levels, membrane lipid rafts abundance and CD11b expression. In parallel experiments, when TNF-α and IL-8, instead of LPS, was used for cell stimulation, L-4F and/or apoA-I revealed only limited efficacy. In LPS-stimulated leukocytes, L-4F was as effective as apoA-I in reducing superoxide formation in 50 % donor plasma, and more effective in 5 % donor plasma (P<0.05). Limulus ambocyte lysate (LAL) and surface plasmon resonance assays showed that L-4F neutralizes LAL endotoxin activity more effectively than apoA-I (P<0.05) likely due to avid binding to LPS. We conclude that (1) direct binding/neutralization of LPS is a major mechanism of L-4F in vitro; (2) while L-4F has similar efficacy to apoA-I in anti-endotoxin effects in whole blood, it demonstrates superior efficacy to apoA-I in aqueous solutions and fluids with limited plasma components. This study rationalizes the utility of L-4F in the treatment of inflammation that is mediated by endotoxin-activated neutrophils.


Subject(s)
Apolipoprotein A-I/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Molecular Mimicry/drug effects , Neutrophils/drug effects , Neutrophils/metabolism , Peptides/pharmacology , Amino Acid Sequence , Dose-Response Relationship, Drug , Humans , Lipopolysaccharides/toxicity , Molecular Mimicry/physiology , Molecular Sequence Data , Peptides/genetics
7.
Innate Immun ; 20(8): 867-80, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24323453

ABSTRACT

Endotoxemia is a major cause of chronic inflammation, and is an important pathogenic factor in the development of metabolic syndrome and atherosclerosis. Human apolipoprotein E (apoE) and apoA-I are protein components of high-density lipoprotein, which have strong anti-endotoxin activity. Here, we compared anti-endotoxin activity of Ac-hE18A-NH2 and 4F peptides, modified from model amphipathic helical 18A peptide, to mimic, respectively, apoE and apoA-I properties. Ac-hE18A-NH2, stronger than 4F, inhibited endotoxin activity and disaggregated Escherichia coli 055:B5 (wild smooth serotype). Ac-hE18A-NH2 and 4F inhibited endotoxin activity of E. coli 026:B6 (rough-like serotype) to a similar degree. This suggests that Ac-hE18A-NH2 as a dual-domain molecule might interact with both the lipid A and headgroup of smooth LPS, whereas 4F binds lipid A. In C57BL/6 mice, Ac-hE18A-NH2 was superior to 4F in inhibiting the inflammatory responses mediated by E. coli 055:B5, but not E. coli 026:B6. However, in THP-1 cells, isolated human primary leukocytes, and whole human blood, Ac-hE18A-NH2 reduced responses more strongly than 4F to both E. coli serotypes either when peptides were pre-incubated or co-incubated with LPS, indicating that Ac-hE18A-NH2 also has strong anti-inflammatory effects independent of endotoxin-neutralizing properties. In conclusion, Ac-hE18A-NH2 is more effective than 4F in inhibiting LPS-mediated inflammation, which opens prospective clinical applications for Ac-hE18A-NH2.


Subject(s)
Apolipoproteins A/pharmacology , Apolipoproteins E/pharmacology , Endotoxins/antagonists & inhibitors , Peptides/pharmacology , Amino Acid Sequence , Animals , Biomimetics , Cell Line , Female , Humans , In Vitro Techniques , Leukocytes/drug effects , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Molecular Sequence Data
8.
Biochem Biophys Res Commun ; 436(4): 705-10, 2013 Jul 12.
Article in English | MEDLINE | ID: mdl-23791744

ABSTRACT

The cationic single domain peptide mR18L has demonstrated lipid-lowering and anti-atherogenic properties in different dyslipidemic mouse models. Lipopolysaccharide (LPS)-mediated inflammation is considered as one of the potential triggers for atherosclerosis. Here, we evaluated anti-inflammatory effects of mR18L peptide against LPS-mediated inflammation. First, we tested the efficacy and tolerance of 1, 2.5 and 5mg/kg mR18L in normolipidemic rats stimulated with 5mg/kg LPS. LPS and then mR18L were injected in different intraperitoneal regions. By 2h post LPS, mR18L inhibited LPS-mediated plasma TNF-α elevation at all doses, with the effect being stronger for 2.5mg/kg (P<0.05 vs. 1mg/kg, non-significant vs. 5mg/kg). In a similar model, 2.5mg/kg mR18L reduced LPS-mediated inflammation in the liver, as assessed by microscopic examination of liver sections and measurements of iNOS expression in the liver tissue. In plasma, 2.5mg/kg mR18L decreased levels of TNF-α and IL-6, decreased endotoxin activity and enhanced HDL binding to LPS. In another similar experiment, mR18L administered 1h post LPS, prevented elevation of plasma triglycerides by 6h post LPS and increased plasma activity of anti-oxidant enzyme paraoxonase 1, along with noted trends in reducing plasma levels of endotoxin and IL-6. Surface plasmon resonance study revealed that mR18L readily binds LPS. We conclude that mR18L exerts anti-endotoxin activity at least in part due to direct LPS-binding and LPS-neutralizing effects. We suggest that anti-endotoxin activity of mR18L is an important anti-inflammatory property, which may increase anti-atherogenic potential of this promising orally active lipid-lowering peptide.


Subject(s)
Hypolipidemic Agents/pharmacology , Inflammation/prevention & control , Lipids/blood , Lipopolysaccharides/toxicity , Liver/drug effects , Peptides/pharmacology , Animals , Cations , Inflammation/chemically induced , Liver/pathology , Rats , Surface Plasmon Resonance
9.
PLoS One ; 8(5): e64486, 2013.
Article in English | MEDLINE | ID: mdl-23691230

ABSTRACT

Acute respiratory distress syndrome (ARDS) due to sepsis has a high mortality rate with limited treatment options. High density lipoprotein (HDL) exerts innate protective effects in systemic inflammation. However, its role in ARDS has not been well studied. Peptides such as L-4F mimic the secondary structural features and functions of apolipoprotein (apo)A-I, the major protein component of HDL. We set out to measure changes in HDL in sepsis-mediated ARDS patients, and to study the potential of L-4F to prevent sepsis-mediated ARDS in a rodent model of lipopolysaccharide (LPS)-mediated acute lung injury, and a combination of primary human leukocytes and human ARDS serum. We also analyzed serum from non-lung disease intubated patients (controls) and sepsis-mediated ARDS patients. Compared to controls, ARDS demonstrates increased serum endotoxin and IL-6 levels, and decreased HDL, apoA-I and activity of anti-oxidant HDL-associated paraoxanase-1. L-4F inhibits the activation of isolated human leukocytes and neutrophils by ARDS serum and LPS in vitro. Further, L-4F decreased endotoxin activity and preserved anti-oxidant properties of HDL both in vitro and in vivo. In a rat model of severe endotoxemia, L-4F significantly decreased mortality and reduces lung and liver injury, even when administered 1 hour post LPS. Our study suggests the protective role of the apoA-I mimetic peptide L-4F in ARDS and gram-negative endotoxemia and warrant further clinical evaluation. The main protective mechanisms of L-4F are due to direct inhibition of endotoxin activity and preservation of HDL anti-oxidant activity.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Apolipoprotein A-I/chemistry , Endotoxemia/complications , Peptidomimetics/pharmacology , Respiratory Distress Syndrome/complications , Respiratory Distress Syndrome/drug therapy , Adult , Aged , Amino Acid Sequence , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , Biomarkers/metabolism , Female , Humans , Lipopolysaccharides/pharmacology , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Middle Aged , Neutrophil Activation/drug effects , Neutrophils/drug effects , Neutrophils/immunology , Peptidomimetics/chemistry , Peptidomimetics/therapeutic use , Rats , Respiratory Distress Syndrome/blood , Respiratory Distress Syndrome/immunology , Superoxides/metabolism , Survival Analysis
10.
J Alzheimers Dis ; 36(2): 335-47, 2013.
Article in English | MEDLINE | ID: mdl-23603398

ABSTRACT

BACKGROUND: Apolipoprotein E (ApoE) is the major apolipoprotein present in the high-density lipoprotein-like particles in the central nervous system (CNS). ApoE is involved in various protective functions in CNS including cholesterol transport, anti-inflammatory, and antioxidant effects. An ApoE peptide would be expected to exert protective effects on neuroinflammation. OBJECTIVE: To determine the effects of an ApoE mimetic peptide Ac-hE18A-NH2 on amyloid-ß pathology. METHOD: Using human APP/PS1ΔE9 transgenic mice and in vitro studies, we have evaluated the effect of an ApoE mimetic peptide, Ac-hE18A-NH2, on amyloid plaque deposition and inflammation. RESULTS: Administration of Ac-hE18A-NH2 to APP/PS1ΔE9 mice for 6 weeks (50 µg/mouse, 3 times a week) significantly improved cognition with a concomitant decrease in amyloid plaque deposition and reduced activated microglia and astrocytes, and increased brain ApoE levels. Oligomeric Aß42 (oAß42) and oxidized PAPC (ox-PAPC) inhibited secretion of ApoE in U251 cells, a human astrocyte cell line, and this effect was ameliorated in the presence of peptide Ac-hE18A-NH2. The peptide also increased Aß42 uptake in a cell line of human macrophages. CONCLUSIONS: Peptide Ac-hE18A-NH2 attenuates the effects of oxidative stress on ApoE secretion, inhibits amyloid plaque deposition, and thus could be beneficial in the treatment of Alzheimer's disease.


Subject(s)
Amyloid Neuropathies/drug therapy , Antipsychotic Agents/therapeutic use , Brain/metabolism , Lipoproteins/therapeutic use , Peptide Fragments/therapeutic use , Amyloid Neuropathies/complications , Amyloid Neuropathies/metabolism , Amyloid Neuropathies/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Apolipoproteins E/metabolism , Brain/drug effects , Cell Line, Transformed , Cholesterol/blood , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Cognition Disorders/genetics , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Maze Learning/drug effects , Mice , Mice, Transgenic , Mutation/genetics , Peptide Fragments/metabolism , Plaque, Amyloid/drug therapy , Plaque, Amyloid/etiology , Plaque, Amyloid/genetics , Presenilin-1/genetics , Transfection
11.
Biochem Biophys Res Commun ; 430(3): 975-80, 2013 Jan 18.
Article in English | MEDLINE | ID: mdl-23261466

ABSTRACT

High density lipoprotein (HDL) associated paraoxonase-1 (PON1) is crucial for the anti-oxidant, anti-inflammatory, and anti-atherogenic properties of HDL. Discoidal apolipoprotein (apo)A-I:1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) complex has been shown to be the most effective in binding PON1, stabilizing it, and enhancing its lactonase and inhibitory activity of low density lipoprotein oxidation. Based on our earlier study demonstrating that apoA-I mimetic peptide 4F forms discoidal complex with 1,2-dimyristoyl-sn-glycero-3-phosphocholine, we hypothesized that lipid complexes of 4F would be able to bind PON1 and enhance its activity and stability. To test our hypothesis, we have expressed and purified a recombinant PON1 (rPON1) and studied its interaction with 4F:POPC complex. Our studies show significant increase, compared to the control, in the paraoxonase activity and stability of rPON1 in the presence of 4F:POPC complex. We propose that 4F:POPC complex is a novel platform for PON1 binding, increasing its stability, and enhancing its enzyme activity. We propose a structural model for the 4F:POPC:PON1 ternary complex that is consistent with our results and published observations.


Subject(s)
Apolipoprotein A-I/metabolism , Aryldialkylphosphatase/metabolism , Peptides/metabolism , Phosphatidylcholines/metabolism , Amino Acid Sequence , Apolipoprotein A-I/chemistry , Aryldialkylphosphatase/genetics , Enzyme Stability , Humans , Models, Molecular , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptides/chemistry , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
12.
Atherosclerosis ; 227(1): 58-64, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23159231

ABSTRACT

OBJECTIVE: We investigated two apoE mimetic peptides with similar long-term plasma cholesterol reducing abilities for their effects on atherosclerotic lesions in Western diet-fed female LDL-receptor (LDL-R) null mice. METHODS AND RESULTS: Single doses of peptides Ac-hE18A-NH(2) and mR18L were administered retro-orbitally to LDL-R null mice on Western diet and plasma cholesterol was measured at 10 min, 4 h, and 24 h post administration. Peptide mR18L and not Ac-hE18A-NH(2) reduced plasma cholesterol levels significantly at 4 h post administration. However, multiple administrations (100 µg/mouse twice weekly for 8 weeks) resulted in a similar reduction in plasma cholesterol. Only the plasma from the Ac-hE18A-NH(2) group had significantly reduced reactive oxygen species levels at the end of the treatment protocol. Both mR18L and Ac-hE18A-NH(2) showed reduced atherosclerotic lesion areas. However, peptide Ac-hE18A-NH(2) was significantly more effective in inhibiting atherosclerosis. Both peptides reduced total plaque macrophage load compared to the saline treated animals, with peptide Ac-hE18A-NH(2) having a greater reduction. Incubation of HepG2 cells and THP-1 monocyte-derived macrophages with both peptides in the presence of oxidized phospholipid showed that Ac-hE18A-NH(2) promotes the secretion of apoE from the cells whereas mR18L does not. CONCLUSIONS: Despite similar reductions in plasma cholesterol levels, Ac-hE18A-NH(2) was more effective in inhibiting lesions than mR18L, possibly due to its ability to promote the secretion of apoE from hepatocytes and macrophages.


Subject(s)
Apolipoproteins E/metabolism , Atherosclerosis/prevention & control , Lipoproteins/pharmacology , Peptide Fragments/pharmacology , Peptides/pharmacology , Receptors, LDL/genetics , Animals , Apolipoproteins E/chemistry , Cholesterol/blood , Female , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Macrophages/drug effects , Macrophages/metabolism , Mice , Reactive Oxygen Species/blood
13.
Arterioscler Thromb Vasc Biol ; 32(11): 2631-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22982462

ABSTRACT

OBJECTIVE: The apolipoprotein A-I (apoA-I) mimetic peptide 4F favors the differentiation of human monocytes to an anti-inflammatory phenotype and attenuates lipopolysaccharide (LPS)-induced inflammatory responses. We investigated the effects of LPS on the Toll-like receptor (TLR) signaling pathway in 4F-differentiated monocyte-derived macrophages. METHODS AND RESULTS: Monocyte-derived macrophages were pretreated with 4F or vehicle for 7 days. 4F downregulated cell-surface TLRs (4, 5, and 6) as determined by flow cytometry. 4F attenuated the LPS-dependent upregulation of genes encoding TLR1, 2, and 6 and genes of the MyD88-dependent (CD14, MyD88, TRAF6, interleukin-1 receptor-associated kinase 4, and inhibitor of kappa light polypeptide gene enhancer in B-cells, kinase beta) and MyD88-independent (interferon regulatory factor 3, TANK-binding kinase 1, and Toll-interleukin 1 receptor domain-containing adaptor-inducing interferon-ß) pathways as determined by microarray analysis and quantitative reverse transcriptase polymerase chain reaction. Functional analyses of monocyte-derived macrophages showed that 4F reduced LPS-dependent TLR4 recycling, phosphorylation of nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha, activation and translocation of nuclear factor-κB and inhibited the secretion of tumor necrosis factor-α and interleukin-6 induced by LPS or lipoteichoic acid. These changes were associated with depletion of cellular cholesterol and caveolin, components of membrane lipid rafts. CONCLUSIONS: These data suggest that disruption of rafts by 4F alters the assembly of TLR-ligand complexes in cell membranes and inhibits proinflammatory gene expression in monocyte-derived macrophages, thus attenuating the responsiveness of macrophages to LPS.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Apolipoprotein A-I/pharmacology , Inflammation/prevention & control , Macrophages/drug effects , Peptides/pharmacology , Toll-Like Receptors/drug effects , Active Transport, Cell Nucleus , Caveolin 1/metabolism , Cells, Cultured , Cholesterol/metabolism , Cytokines/genetics , Cytokines/metabolism , Down-Regulation , Flow Cytometry , Gene Expression Profiling/methods , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Macrophages/immunology , Membrane Microdomains/drug effects , Membrane Microdomains/immunology , Oligonucleotide Array Sequence Analysis , Phosphorylation , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Time Factors , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Transcription, Genetic/drug effects
14.
Atherosclerosis ; 224(2): 326-31, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22771190

ABSTRACT

OBJECTIVE: The apolipoprotein E mimetic peptide Ac-hE18A-NH(2), capable of reducing plasma cholesterol and possessing anti-inflammatory properties, was compared with the well-studied anti-atherogenic apoA-I mimetic peptide 4F for reducing lesion formation in female apoE null mice with already existing lesions. METHODS AND RESULTS: In initial experiments, Ac-hE18A-NH(2) was administered retro-orbitally two or three times weekly for 6-8 weeks, while peptide 4F was administered intraperitoneally every day for the same period. Age matched controls were injected with saline every day. At the end of the treatment period, plasma cholesterol levels of Ac-hE18A-NH(2) administered mice were significantly lower than in 4F and control mice. However, both 4F and Ac-hE18A-NH(2) showed reduced lesion areas in en face lesion analysis to a similar extent compared to the control group, while paraoxonase-1 (PON-1) activity was increased only in the Ac-hE18A-NH(2) group. In the third experiment, both peptides were administered at the same dose, frequency, and route of administration. The reduction in en face lesions with Ac-hE18A-NH(2) was significantly greater than the 4F and control groups, although lesions in 4F-treated mice were also significantly reduced compared with controls. Both peptide groups had significantly reduced plasma lipid hydroperoxides, but only the Ac-hE18A-NH(2) group had significantly reduced serum amyloid A levels. HDL and plasma inflammatory indices were significantly reduced in both peptide groups compared with controls. CONCLUSIONS: Although both peptides had similar anti-inflammatory properties, Ac-hE18A-NH(2) was more effective in inhibiting lesions than 4F at the same dose, frequency, and route of administration, perhaps due to its cholesterol reducing properties.


Subject(s)
Anticholesteremic Agents/pharmacology , Aortic Diseases/prevention & control , Apolipoprotein A-I/pharmacology , Apolipoproteins E/deficiency , Atherosclerosis/prevention & control , Lipoproteins/pharmacology , Peptide Fragments/pharmacology , Age Factors , Aging , Animals , Anti-Inflammatory Agents/pharmacology , Anticholesteremic Agents/administration & dosage , Antioxidants/pharmacology , Aortic Diseases/blood , Aortic Diseases/genetics , Aortic Diseases/pathology , Apolipoprotein A-I/administration & dosage , Apolipoproteins E/genetics , Aryldialkylphosphatase/blood , Atherosclerosis/blood , Atherosclerosis/genetics , Atherosclerosis/pathology , Cholesterol/blood , Disease Models, Animal , Drug Administration Schedule , Female , Injections, Intraperitoneal , Injections, Intravenous , Lipid Peroxides/blood , Lipoproteins/administration & dosage , Mice , Mice, Knockout , Peptide Fragments/administration & dosage , Serum Amyloid A Protein/metabolism , Sex Factors , Time Factors
15.
J Lipid Res ; 53(8): 1576-87, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22589558

ABSTRACT

Myeloperoxidase (MPO)-derived hypochlorous acid induces changes in HDL function via redox modifications at the level of apolipoprotein A-I (apoA-I). As 4F and apoA-I share structural and functional properties, we tested the hypothesis that 4F acts as a reactive substrate for hypochlorous acid (HOCl). 4F reduced the HOCl-mediated oxidation of the fluorescent substrate APF in a concentration-dependent manner (ED(50) ∼ 56 ± 3 µM). This reaction induced changes in the physical properties of 4F. Addition of HOCl to 4F at molar ratios ranging from 1:1 to 3:1 reduced 4F band intensity on SDS-PAGE gels and was accompanied by the formation of a higher molecular weight species. Chromatographic studies showed a reduction in 4F peak area with increasing HOCl and the formation of new products. Mass spectral analyses of collected fractions revealed oxidation of the sole tryptophan (Trp) residue in 4F. 4F was equally susceptible to oxidation in the lipid-free and lipid-bound states. To determine whether Trp oxidation influenced its apoA-I mimetic properties, we monitored effects of HOCl on 4F-mediated lipid binding and ABCA1-dependent cholesterol efflux. Neither property was altered by HOCl. These results suggest that 4F serves as a reactive substrate for HOCl, an antioxidant response that does not influence the lipid binding and cholesterol effluxing capacities of the peptide.


Subject(s)
Apolipoprotein A-I/chemistry , Peptides/chemistry , Peptides/metabolism , Peptidomimetics/chemistry , Peptidomimetics/metabolism , Amino Acid Sequence , Biological Transport/drug effects , Cell Line, Tumor , Cholesterol/metabolism , Humans , Hypochlorous Acid/metabolism , Hypochlorous Acid/pharmacology , Molecular Dynamics Simulation , Molecular Sequence Data , Oxidation-Reduction/drug effects , Protein Conformation
16.
J Lipid Res ; 53(5): 849-858, 2012 May.
Article in English | MEDLINE | ID: mdl-22377531

ABSTRACT

To test the hypothesis that sidedness of interfacial arginine (Arg) in apoA-I mimetic peptides, similar to that observed in apoA-I (Bashtovyy, D. et al. 2011. Sequence conservation of apolipoprotein A-I affords novel insights into HDL structure-function. J. Lipid Res. 52: 435-450.), may be important for biological activity, we compared properties of 4F and analogs, [K4,¹5>R]4F and [K9,¹³>R]4F, with Lys>Arg substitutions on the right and left side, respectively, of the 4F amphipathic helix. Intraperitoneal administration of these peptides into female apoE null mice (n = 13 in each group) reduced en face lesions significantly compared with controls; 4F and [K4,¹5>R]4F were equally effective whereas [K9,¹³>R]4F was less effective. Turnover experiments indicated that [K4,¹5>R]4F reached the highest, whereas [K9,¹³>R]4F had the lowest, plasma peak levels with a similar half life as the [K4,¹5>R]4F analog. The half life of 4F was two times longer than the other two peptides. The order in their abilities to associate with HDL in human plasma, generation of apoA-I particles with pre-ß mobility from isolated HDL, lipid associating ability, and sensitivity of lipid complexes to trypsin digestion was: 4F>[K4,¹5,>R]4F>[K9,¹³>R]4F. These studies support our hypothesis that the sidedness of interfacial Arg residues in the polar face of apoA-I mimetics results in differential biological properties.


Subject(s)
Apolipoprotein A-I/chemistry , Arginine/chemistry , Atherosclerosis/drug therapy , Peptidomimetics/chemistry , Peptidomimetics/pharmacology , Animals , Apolipoprotein A-I/deficiency , Apolipoprotein A-I/genetics , Aryldialkylphosphatase/metabolism , Atherosclerosis/blood , Atherosclerosis/metabolism , Chemical Phenomena , Cholesterol/blood , Female , Gene Deletion , Guanidine/pharmacology , Humans , Lipoproteins, HDL/metabolism , Mice , Oxidation-Reduction , Peptidomimetics/metabolism , Peptidomimetics/therapeutic use , Phosphatidylcholines/metabolism , Protein Structure, Secondary , Protein Unfolding/drug effects , Reactive Oxygen Species/blood , Unilamellar Liposomes/metabolism
17.
Biochim Biophys Acta ; 1808(1): 498-507, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20970404

ABSTRACT

The surprising observation that a 10-residue class G(⁎) peptide from apolipoprotein J, [113-122]apoJ, possesses anti-inflammatory and anti-atherogenic properties prompted us to delineate its structural characteristics in the presence of normal and oxidized lipid. Towards this, we have determined high-resolution structure of [113-122]apoJ in solution using nuclear magnetic resonance (NMR) spectroscopy and studied its interaction with lipids, including oxidized lipids, using a number of biophysical methods. Circular dichroism and NMR studies established that in the presence of dodecylphosphocholine (DPC) micelle, this peptide adopts amphipathic α-helical structure. The observed Nuclear Overhauser effects indicate that the amphipathic helical structure of the peptide is stabilized by the N-terminal acetyl and C-terminal amide blocking groups. We used isothermal titration calorimetry to measure binding enthalpy of the peptide with DPC micelle, an oxidized lipid, 1-(palmitoyl)-2-(5-keto-6-octene-dioyl) phosphatidylcholine (KOdiA-PC), and the mixture of these two lipids (5mol% KOdiA-PC in DPC micelle). We find that the peptide binding with DPC micelle is associated with an enthalpy change (-16.75±0.16 Kcal/mol) much larger than that resulting from the binding with KodiA-PC (-3.67±0.13 Kcal/mol). Incorporation of a small amount of KOdiA-PC (5mol%) in DPC micelle also results in the lowering of peptide binding enthalpy (-13.43±0.18 Kcal/mol). These results are consistent with overall negative charge and altered conformational properties of oxidized sn-2 chain of KOdiA-PC. Our results have unambiguously established the amphipathic α-helical structure of [113-122]apoJ peptide in the presence of DPC micelle as well as its ability to bind oxidized lipid. These in vitro results help explain the previously observed anti-inflammatory and anti-atherosclerotic properties of this peptide.


Subject(s)
Clusterin/chemistry , Lipids/chemistry , Magnetic Resonance Spectroscopy/methods , Anti-Inflammatory Agents/pharmacology , Biophysics/methods , Calorimetry/methods , Chemistry, Pharmaceutical/methods , Circular Dichroism/methods , Micelles , Peptides/chemistry , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/chemistry , Protein Binding , Protein Conformation , Protein Structure, Secondary , Thermodynamics
18.
Atherosclerosis ; 213(2): 449-57, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21030022

ABSTRACT

OBJECTIVE: We recently described anti-atherogenic properties of the dual domain peptide Ac-hE18A-NH(2) derived by covalently linking the heparin binding domain 141-150 of apoE to 18A, a class A amphipathic helical peptide. In this paper we have compared the properties of Ac-hE18A-NH(2) with the non-heparin binding 151-160 region of apoE linked to 18A (Ac-nhE18A-NH(2)). METHODS AND RESULTS: Both peptides were highly helical in solution and in association with lipids. Ac-hE18A-NH(2) and not Ac-nhE18A-NH(2) enhanced uptake of low density lipoprotein (LDL) in HepG2 cells. While Ac-hE18A-NH(2) retarded the electrophoretic mobility of LDL, Ac-nhE18A-NH(2) slightly enhanced mobility. Ac-hE18A-NH(2) reduced monocyte association with endothelial cells, while Ac-nhE18A-NH(2) increased it. Ac-hE18A-NH(2) also reduced lipid hydroperoxide content of LDL while Ac-nhE18A-NH(2) increased it. A single administration of Ac-hE18A-NH(2) (100 µg/mouse) into apoE null mice dramatically reduced cholesterol (from 600 mg/dL to 180 mg/dL at 5 min and to 60 mg/dL at 5h) while Ac-nhE18A-NH(2) had no effect. Administration (100 µg/mouse/day, three days a week) into apoE null mice for six weeks showed Ac-hE18A-NH(2) group having a moderate aortic sinus lesion reduction compared with the control group (-15.1%), while the Ac-nhE18A-NH(2) administered group had increased lesion area (+33.0% vs controls and 36.1% vs Ac-hE18A-NH(2)). Plasma from mice administered Ac-hE18A-NH(2) for six weeks showed a significant reduction in plasma cholesterol and triglyceride levels and increase in paraoxonase-1 (PON-1) activity compared to controls, while Ac-nhE18A-NH(2) caused no change in plasma cholesterol and decreased PON-1 activity. CONCLUSION: It is proposed that Ac-hE18A-NH(2) reduced lesion progression in apoE null mice due to its anti-inflammatory and lipoprotein clearing properties, while Ac-nhE18A-NH(2) exhibited pro-atherogenic effects.


Subject(s)
Apolipoproteins E/genetics , Atherosclerosis/etiology , Endothelial Cells/drug effects , Lipoproteins/pharmacology , Monocytes/drug effects , Peptide Fragments/pharmacology , Animals , Apolipoproteins E/chemistry , Apolipoproteins E/deficiency , Aryldialkylphosphatase/metabolism , Cholesterol/blood , Female , Hep G2 Cells , Heparan Sulfate Proteoglycans/metabolism , Humans , Lipoproteins, LDL/drug effects , Lipoproteins, LDL/metabolism , Mice , Protein Structure, Secondary , Protein Structure, Tertiary , Triglycerides/blood
19.
J Lipid Res ; 51(12): 3491-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20841495

ABSTRACT

We have shown that Ac-hE18A-NH2, a dual-domain cationic apolipoprotein-mimetic peptide, reduces plasma cholesterol levels in dyslipidemic mice. Two single-domain cationic peptides based on the lytic class L peptide 18L were developed to test the hypothesis that a single-domain cationic amphipathic peptide can reduce atherosclerosis in apolipoprotein (apo)E null mice when orally administered. To incorporate anti-inflammatory properties, aromatic residues were clustered in the nonpolar face similar to peptide 4F, resulting in modified 18L (m18L). To reduce lytic properties, the Lys residues of 18L were replaced with Arg with the resulting peptide called modified R18L (mR18L). Biophysical studies showed that mR18L had stronger interactions with lipids than did m18L. Peptide mR18L was also more effective than m18L in promoting LDL uptake by HepG2 cells. ApoE null mice received normal chow or chow containing m18L or mR18L for six weeks. A significant reduction in plasma cholesterol and aortic sinus lesion area was seen only in the mR18L group. Plasma from mice administered mR18L, unlike those from the control and m18L groups, did not enhance monocyte adhesion to endothelial cells. Thus oral administration of mR18L reduces plasma cholesterol and lesion formation and inhibits monocyte adhesion.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Aortic Aneurysm/drug therapy , Atherosclerosis/drug therapy , Endothelial Cells/drug effects , Monocytes/drug effects , Myelin Basic Protein/therapeutic use , Peptide Fragments/therapeutic use , Peptides/therapeutic use , Administration, Oral , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology , Aortic Aneurysm/metabolism , Aortic Aneurysm/pathology , Arginine/chemistry , Arginine/metabolism , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cations , Cell Adhesion/drug effects , Cholesterol/blood , Cholesterol/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Hep G2 Cells , Humans , Lipid Metabolism/drug effects , Lysine/chemistry , Lysine/metabolism , Mice , Mice, Knockout , Monocytes/metabolism , Monocytes/pathology , Myelin Basic Protein/administration & dosage , Myelin Basic Protein/pharmacology , Peptide Fragments/administration & dosage , Peptide Fragments/pharmacology , Peptides/administration & dosage , Peptides/pharmacology , Protein Structure, Secondary , Protein Structure, Tertiary
20.
Adv Exp Med Biol ; 660: 1-4, 2010.
Article in English | MEDLINE | ID: mdl-20221865

ABSTRACT

ApoE mimetic peptide possesses the putative receptor binding domain 141-150 (LRKLRKRLLR) of apoE covalently linked to the class A amphipathic helical peptide 18A. It dramatically reduces plasma cholesterol in dyslipidemic mouse and rabbit models. Recycling of apoE mimetic peptide increases the duration of preß-HDL formation leading to extended anti-inflammatory and atheroprotective properties.


Subject(s)
Apolipoproteins E/chemistry , Aryldialkylphosphatase/chemistry , Hydrogen Peroxide/chemistry , Lipids/chemistry , Lipoproteins, HDL/chemistry , Animals , Anti-Inflammatory Agents/pharmacology , Atherosclerosis/prevention & control , Cell Line , Humans , Mice , Peptides/chemistry , Rabbits , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...