Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Lipid Res ; 59(11): 2075-2083, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30201630

RESUMO

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.


Assuntos
Apolipoproteínas E/química , Colesterol/sangue , Peptídeos/química , Peptídeos/farmacologia , Animais , LDL-Colesterol/sangue , Cromatografia Líquida de Alta Pressão , Eletroforese em Gel de Ágar , Feminino , Haplorrinos , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Metabolismo dos Lipídeos/efeitos dos fármacos , Macaca , Masculino , Camundongos , Camundongos Knockout , Peptídeos/sangue
2.
Adv Exp Med Biol ; 982: 407-429, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28551800

RESUMO

Lipoproteins play a key role in regulating plasma and tissue levels of cholesterol. Apolipoprotein B (apoB)-containing lipoproteins, including chylomicrons, very-low density lipoprotein (VLDL) and low-density lipoprotein (LDL), serve as carriers of triglycerides and cholesterol and deliver these metabolites to peripheral tissues. In contrast, high-density lipoprotein (HDL) mediates Reverse Cholesterol Transport (RCT), a process by which excess cholesterol is removed from the periphery and taken up by hepatocytes where it is metabolized and excreted. Anti-atherogenic properties of HDL have been largely ascribed to apoA-I, the major protein component of the lipoprotein particle. The inflammatory response associated with atherosclerosis and ischemia-reperfusion (I-R) injury has been linked to the development of mitochondrial dysfunction. Under these conditions, an increase in reactive oxygen species (ROS) formation induces damage to mitochondrial structural elements, leading to a reduction in ATP synthesis and initiation of the apoptotic program. Recent studies suggest that HDL-associated apoA-I and lysosphingolipids attenuate mitochondrial injury by multiple mechanisms, including the suppression of ROS formation and induction of autophagy. Other apolipoproteins, however, present in lower abundance in HDL particles may exert opposing effects on mitochondrial function. This chapter examines the role of HDL-associated apolipoproteins and lipids in the regulation of mitochondrial function and bioenergetics.


Assuntos
Lipoproteínas HDL/metabolismo , Mitocôndrias/metabolismo , Transdução de Sinais , Animais , Apolipoproteína A-I/metabolismo , Autofagia , Humanos , Lisofosfolipídeos/metabolismo , Dinâmica Mitocondrial , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo
3.
Biochem J ; 467(3): 517-27, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25742174

RESUMO

The apoA-I (apolipoprotein A-I) mimetic peptide 4F favours the differentiation of human monocytes to an alternatively activated M2 phenotype. The goal of the present study was to test whether the 4F-mediated differentiation of MDMs (monocyte-derived macrophages) requires the induction of an oxidative metabolic programme. 4F treatment induced several genes in MDMs that play an important role in lipid metabolism, including PPARγ (peroxisome-proliferator-activated receptor γ) and CD36. Addition of 4F was associated with a significant increase in FA (fatty acid) uptake and oxidation compared with vehicle treatment. Mitochondrial respiration was assessed by measurement of the OCR (oxygen-consumption rate). 4F increased basal and ATP-linked OCR as well as maximal uncoupled mitochondrial respiration. These changes were associated with a significant increase in ΔΨm (mitochondrial membrane potential). The increase in metabolic activity in 4F-treated MDMs was attenuated by etomoxir, an inhibitor of mitochondrial FA uptake. Finally, addition of the PPARγ antagonist T0070907 to 4F-treated MDMs reduced the expression of CD163 and CD36, cell-surface markers for M2 macrophages, and reduced basal and ATP-linked OCR. These results support our hypothesis that the 4F-mediated differentiation of MDMs to an anti-inflammatory phenotype is due, in part, to an increase in FA uptake and mitochondrial oxidative metabolism.


Assuntos
Apolipoproteína A-I/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Peptídeos/farmacologia , Anti-Inflamatórios/farmacologia , Benzamidas/farmacologia , Materiais Biomiméticos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Células Cultivadas , Metabolismo Energético , Ácidos Graxos/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/genética , Macrófagos/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Monócitos/citologia , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Consumo de Oxigênio , PPAR gama/antagonistas & inibidores , Piridinas/farmacologia
4.
Arterioscler Thromb Vasc Biol ; 32(11): 2631-9, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22982462

RESUMO

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.


Assuntos
Anti-Inflamatórios/farmacologia , Apolipoproteína A-I/farmacologia , Inflamação/prevenção & controle , Macrófagos/efeitos dos fármacos , Peptídeos/farmacologia , Receptores Toll-Like/efeitos dos fármacos , Transporte Ativo do Núcleo Celular , Caveolina 1/metabolismo , Células Cultivadas , Colesterol/metabolismo , Citocinas/genética , Citocinas/metabolismo , Regulação para Baixo , Citometria de Fluxo , Perfilação da Expressão Gênica/métodos , Humanos , Inflamação/genética , Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/imunologia , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/imunologia , Análise de Sequência com Séries de Oligonucleotídeos , Fosforilação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo , Transcrição Gênica/efeitos dos fármacos
5.
J Lipid Res ; 53(8): 1576-87, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22589558

RESUMO

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.


Assuntos
Apolipoproteína A-I/química , Peptídeos/química , Peptídeos/metabolismo , Peptidomiméticos/química , Peptidomiméticos/metabolismo , Sequência de Aminoácidos , Transporte Biológico/efeitos dos fármacos , Linhagem Celular Tumoral , Colesterol/metabolismo , Humanos , Ácido Hipocloroso/metabolismo , Ácido Hipocloroso/farmacologia , Simulação de Dinâmica Molecular , Dados de Sequência Molecular , Oxirredução/efeitos dos fármacos , Conformação Proteica
6.
Am J Physiol Cell Physiol ; 298(6): C1538-48, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20219948

RESUMO

HDL and its major protein component apolipoprotein A-I (apoA-I) exert anti-inflammatory effects, inhibit monocyte chemotaxis/adhesion, and reduce vascular macrophage content in inflammatory conditions. In this study, we tested the hypothesis that the apoA-I mimetic 4F modulates the function of monocyte-derived macrophages (MDMs) by regulating the expression of key cell surface receptors on MDMs. Primary human monocytes and THP-1 cells were treated with 4F, apoA-I, or vehicle for 7 days and analyzed for expression of cell surface markers, adhesion to human endothelial cells, phagocytic function, cholesterol efflux capacity, and lipid raft organization. 4F and apoA-I treatment decreased the expression of HLA-DR, CD86, CD11b, CD11c, CD14, and Toll-like receptor-4 (TLR-4) compared with control cells, suggesting the induction of monocyte differentiation. Both treatments abolished LPS-induced mRNA for monocyte chemotactic protein-1 (MCP-1), macrophage inflammatory protein-1 (MIP-1), regulated on activation, normal T-expressed and presumably secreted (RANTES), IL-6, and TNF-alpha but significantly upregulated LPS-induced IL-10 expression. Moreover, 4F and apoA-I induced a 90% reduction in the expression of CD49d, a ligand for the VCAM-1 receptor, with a concurrent decrease in monocyte adhesion (55% reduction) to human endothelial cells and transendothelial migration (34 and 27% for 4F and apoA-I treatments) compared with vehicle treatment. In addition, phagocytosis of dextran-FITC beads was inhibited by 4F and apoA-I, a response associated with reduced expression of CD32. Finally, 4F and apoA-I stimulated cholesterol efflux from MDMs, leading to cholesterol depletion and disruption of lipid rafts. These data provide evidence that 4F, similar to apoA-I, induces profound functional changes in MDMs, possibly due to differentiation to an anti-inflammatory phenotype.


Assuntos
Anti-Inflamatórios/farmacologia , Apolipoproteína A-I/farmacologia , Macrófagos/efeitos dos fármacos , Antígenos CD/metabolismo , Adesão Celular/efeitos dos fármacos , Moléculas de Adesão Celular/metabolismo , Células Cultivadas , Colesterol/metabolismo , Citocinas/genética , Citocinas/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Antígenos HLA-DR/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Migração e Rolagem de Leucócitos/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Macrófagos/imunologia , Macrófagos/metabolismo , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Mimetismo Molecular , Fagocitose/efeitos dos fármacos , Fenótipo , RNA Mensageiro/metabolismo
7.
J Lipid Res ; 51(12): 3491-9, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20841495

RESUMO

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.


Assuntos
Anti-Inflamatórios/uso terapêutico , Aneurisma Aórtico/tratamento farmacológico , Aterosclerose/tratamento farmacológico , Células Endoteliais/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Proteína Básica da Mielina/uso terapêutico , Fragmentos de Peptídeos/uso terapêutico , Peptídeos/uso terapêutico , Administração Oral , Animais , Anti-Inflamatórios/administração & dosagem , Anti-Inflamatórios/farmacologia , Aneurisma Aórtico/metabolismo , Aneurisma Aórtico/patologia , Arginina/química , Arginina/metabolismo , Aterosclerose/metabolismo , Aterosclerose/patologia , Cátions , Adesão Celular/efeitos dos fármacos , Colesterol/sangue , Colesterol/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Células Hep G2 , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Lisina/química , Lisina/metabolismo , Camundongos , Camundongos Knockout , Monócitos/metabolismo , Monócitos/patologia , Proteína Básica da Mielina/administração & dosagem , Proteína Básica da Mielina/farmacologia , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/farmacologia , Peptídeos/administração & dosagem , Peptídeos/farmacologia , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
8.
J Lipid Res ; 51(9): 2695-705, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20495214

RESUMO

High density lipoprotein (HDL) and apolipoprotein A-I (apoA-I) reduce inflammatory responses to lipopolysaccharide (LPS). We tested the hypothesis that the apoA-I mimetic peptide 4F prevents LPS-induced defects in blood pressure and vascular reactivity. Systolic blood pressure (SBP) was measured in rats at baseline and 6 h after injection of LPS (10 mg/kg) or saline vehicle. Subgroups of LPS-treated rats also received 4F (10 mg/kg) or scrambled 4F (Sc-4F). LPS administration reduced SBP by 35% compared with baseline. 4F attenuated the reduction in SBP in LPS-treated rats (17% reduction), while Sc-4F was without effect. Ex vivo studies showed a reduced contractile response to phenylephrine (PE) in aortae of LPS-treated rats (ED(50) = 459 +/- 83 nM) compared with controls (ED(50) = 57 +/- 6 nM). This was associated with nitric oxide synthase 2 (NOS2) upregulation. 4F administration improved vascular contractility (ED(50) = 60 +/- 9 nM), reduced aortic NOS2 protein, normalized plasma levels of NO metabolites, and reduced mortality in LPS-treated rats. These changes were associated with a reduction in plasma endotoxin activity. In vivo administration of (14)C-4F and Bodipy-LPS resulted in their colocalization and retention in the HDL fraction. It is proposed that 4F promotes the localization of LPS to the HDL fraction, resulting in endotoxin neutralization. 4F may thus prevent LPS-induced hemodynamic changes associated with NOS2 induction.


Assuntos
Apolipoproteína A-I/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Vasos Sanguíneos , Endotoxemia/tratamento farmacológico , Endotoxemia/fisiopatologia , Peptídeos/farmacologia , Animais , Apolipoproteína A-I/genética , Pressão Sanguínea/fisiologia , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/fisiologia , Vasos Sanguíneos/fisiopatologia , HDL-Colesterol/sangue , Inibidores de Ciclo-Oxigenase/farmacologia , Endotoxemia/induzido quimicamente , Hemodinâmica/efeitos dos fármacos , Humanos , Indometacina/farmacologia , Lipopolissacarídeos/farmacologia , Masculino , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/metabolismo , Nitrobenzenos/farmacologia , Peptídeos/genética , Fenilefrina/farmacologia , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Sulfonamidas/farmacologia , Vasoconstritores/farmacologia
9.
Adv Exp Med Biol ; 660: 1-4, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20221865

RESUMO

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.


Assuntos
Apolipoproteínas E/química , Arildialquilfosfatase/química , Peróxido de Hidrogênio/química , Lipídeos/química , Lipoproteínas HDL/química , Animais , Anti-Inflamatórios/farmacologia , Aterosclerose/prevenção & controle , Linhagem Celular , Humanos , Camundongos , Peptídeos/química , Coelhos , Fatores de Tempo
10.
Am J Physiol Gastrointest Liver Physiol ; 297(1): G1-G10, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19443732

RESUMO

Macrophages are first seen in the fetal intestine at 11-12 wk and rapidly increase in number during the 12- to 22-wk period of gestation. The development of macrophage populations in the fetal intestine precedes the appearance of lymphocytes and neutrophils and does not require the presence of dietary or microbial antigens. In this study, we investigated the role of chemerin, a recently discovered, relatively selective chemoattractant for macrophages, in the recruitment of macrophage precursors to the fetal intestine. Chemerin mRNA/protein expression was measured in jejunoileal tissue from 10- to 24-wk human fetuses, neonates operated for intestinal obstruction, and adults undergoing bariatric surgery. The expression of chemerin in intestinal epithelial cells (IECs) was confirmed by using cultured primary IECs and IEC-like cell lines in vitro. The regulatory mechanisms involved in chemerin expression were investigated by in silico and immunolocalization techniques. IECs in the fetal, but not mature, intestine express chemerin. Chemerin expression peaked in the fetal intestine at 20-24 wk and then decreased to original low levels by full term. During the 10- to 24-wk period, chemerin accounted for most of the macrophage chemotactic activity of cultured fetal IECs. The maturational changes in chemerin expression correlated with the expression of retinoic acid receptor-beta in the intestine. Chemerin is an important mediator of epithelial-macrophage cross talk in the fetal/premature, but not in the mature, intestine. Understanding the regulation of the gut macrophage pool is an important step in development of novel strategies to boost mucosal immunity in premature infants and other patient populations at risk of microbial translocation.


Assuntos
Quimiocinas/metabolismo , Quimiotaxia , Células Epiteliais/imunologia , Íleo/imunologia , Jejuno/imunologia , Macrófagos/imunologia , Adulto , Sequência de Aminoácidos , Células CACO-2 , Quimiocinas/genética , Meios de Cultivo Condicionados/metabolismo , Feto/imunologia , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Humanos , Íleo/embriologia , Imuno-Histoquímica , Recém-Nascido , Peptídeos e Proteínas de Sinalização Intercelular , Jejuno/embriologia , Dados de Sequência Molecular , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Receptores do Ácido Retinoico/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Am J Physiol Heart Circ Physiol ; 297(2): H866-73, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19561306

RESUMO

Systemic inflammation induces a multiple organ dysfunction syndrome that contributes to morbidity and mortality in septic patients. Since increasing plasma apolipoprotein A-I (apoA-I) and HDL may reduce the complications of sepsis, we tested the hypothesis that the apoA-I mimetic peptide 4F confers similar protective effects in rats undergoing cecal ligation and puncture (CLP) injury. Male Sprague-Dawley rats were randomized to undergo CLP or sham surgery. IL-6 levels were significantly elevated in plasma by 6 h after CLP surgery compared with shams. In subsequent studies, CLP rats were further subdivided to receive vehicle or 4F (10 mg/kg) by intraperitoneal injection, 6 h after sepsis induction. Sham-operated rats received saline. Echocardiographic studies showed a reduction in left ventricular end-diastolic volume, stroke volume, and cardiac output (CO) 24 h after CLP surgery. These changes were associated with reduced blood volume and left ventricular filling pressure. 4F treatment improved blood volume status, increased CO, and reduced plasma IL-6 in CLP rats. Total cholesterol (TC) and HDL were 79 +/- 5 and 61 +/- 4 mg/dl, respectively, in sham rats. TC was significantly reduced in CLP rats (54 +/- 3 mg/dl) due to a reduction in HDL (26 +/- 3 mg/dl). 4F administration to CLP rats attenuated the reduction in TC (69 +/- 4 mg/dl) and HDL (41 +/- 3 mg/dl) and prevented sepsis-induced changes in HDL protein composition. Increased plasma HDL in 4F-treated CLP rats was associated with an improvement in CO and reduced mortality. It is proposed that protective effects of 4F are related to its ability to prevent the sepsis-induced reduction in plasma HDL.


Assuntos
Apolipoproteína A-I/imunologia , Inflamação , Peptídeos/farmacologia , Sepse , Função Ventricular Esquerda/efeitos dos fármacos , Função Ventricular Esquerda/imunologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/imunologia , Volume Sanguíneo/efeitos dos fármacos , Volume Sanguíneo/imunologia , Ecocardiografia , Inflamação/tratamento farmacológico , Inflamação/imunologia , Inflamação/mortalidade , Interleucina-6/sangue , Lipoproteínas HDL/sangue , Masculino , Mimetismo Molecular/imunologia , Peptídeos/imunologia , Ratos , Ratos Sprague-Dawley , Sepse/tratamento farmacológico , Sepse/imunologia , Sepse/mortalidade , Volume Sistólico/efeitos dos fármacos , Volume Sistólico/imunologia
12.
Vasc Dis Prev ; 6: 122-130, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20084185

RESUMO

Anti-atherogenic effects of high density lipoprotein (HDL) and its major protein component apolipoprotein A-I (apoA-I) are principally thought to be due to their ability to mediate reverse cholesterol transport. These agents also possess anti-oxidant properties that prevent the oxidative modification of low density lipoprotein (LDL) and anti-inflammatory properties that include inhibition of endothelial cell adhesion molecule expression. Results of the Framingham study revealed that a reduction in HDL levels is an independent risk factor for coronary artery disease (CAD). Accordingly, there has been considerable interest in developing new therapies that specifically elevate HDL cholesterol. However, recent evidence suggests that increasing circulating HDL cholesterol levels alone is not sufficient as a mode of HDL therapy. Rather, therapeutic approaches that increase the functional properties of HDL may be superior to simply raising the levels of HDL per se. Our laboratory has pioneered the development of synthetic, apolipoprotein mimetic peptides which are structurally and functionally similar to apoA-I but possess unique structural homology to the lipid-associating domains of apoA-I. The apoA-I mimetic peptide 4F inhibits atherogenic lesion formation in murine models of atherosclerosis. This effect is related to the ability of 4F to induce the formation of pre-ß HDL particles that are enriched in apoA-I and paraoxonase. 4F also possesses anti-inflammatory and anti-oxidant properties that are independent of its effect on HDL quality per se. Recent studies suggest that 4F stimulates the expression of the antioxidant enzymes heme oxygenase and superoxide dismutase and inhibits superoxide anion formation in blood vessels of diabetic, hypercholesterolemic and sickle cell disease mice. The goal of this review is to discuss HDL-dependent and -independent mechanisms by which apoA-I mimetic peptides reduce vascular injury in experimental animal models.

13.
Chem Phys Lipids ; 219: 28-35, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30707910

RESUMO

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.


Assuntos
Apolipoproteína A-I/química , Peptídeos/química , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Sequência de Aminoácidos , Apolipoproteína A-I/análise , Linhagem Celular , Colesterol/metabolismo , Humanos , Ácido Hipocloroso/química , Espectrometria de Massas , Oxirredução , Fosfatidilcolinas/química
14.
Curr Atheroscler Rep ; 10(5): 405-12, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18706282

RESUMO

3-Hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins) are currently the drug of choice for the clinical management of elevated low-density lipoprotein (LDL) cholesterol. Although statin treatment provides an overall improvement in outcomes, clinical trial data reveal a significant number of cardiac events despite reaching targeted LDL levels. A low serum high-density lipoprotein (HDL) cholesterol level is an independent predictor of cardiovascular risk. Accordingly, there has been interest in determining whether HDL elevation, in addition to LDL lowering, further reduces risk in patients with coronary artery disease. Several commonly prescribed lipid-lowering therapies modestly raise HDL, but their use may be limited by the development of adverse reactions. Emerging data suggest that HDL quality and function may also be significantly reduced by atherosclerosis and other inflammatory diseases. The goal of this review is to discuss the current status of HDL therapeutics, with emphasis on a novel class of agent, the apolipoprotein A-I mimetic peptides, which improve the functional properties of HDL cholesterol.


Assuntos
Doenças Cardiovasculares/sangue , Doenças Cardiovasculares/prevenção & controle , HDL-Colesterol/sangue , HDL-Colesterol/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Animais , Apolipoproteína A-I/sangue , Apolipoproteína A-I/farmacologia , Apolipoproteína A-I/uso terapêutico , Aterosclerose/sangue , Aterosclerose/tratamento farmacológico , Proteínas de Transferência de Ésteres de Colesterol/antagonistas & inibidores , Ácido Clofíbrico/farmacologia , Humanos , Lipoproteínas/farmacologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fragmentos de Peptídeos/farmacologia
15.
Curr Top Pept Protein Res ; 19: 15-25, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29955206

RESUMO

Cardiovascular disease, specifically atherosclerosis, is exacerbated by hypercholesterolemia. Current therapies that target lipid lowering, however, are not effective in all patients. Apolipoprotein E (apoE) plays an important role in mediating the clearance of plasma cholesterol and also exerts numerous cytoprotective responses. Our laboratory has synthesized novel therapeutics that mimic the ability of apoE to decrease plasma cholesterol. The apoE mimetic peptide AEM-2 is a dual domain peptide composed of an amphipathic helical region that binds phospholipids and a positively charged region that mediates the hepatic clearance of lipoproteins. Administration of AEM-2 to apoE null mice reduced plasma cholesterol concentration by 80% one hour post-administration. Since apoE is also known to exert anti-inflammatory effects that are independent of its ability to lower cholesterol, we tested effects of AEM-2 on lipopolysaccharide-induced responses in human THP-1 macrophages. Pre-treatment of THP-1 cells with AEM-2 significantly reduced the LPS-induced secretion of IL-6 and TNFα. Since LPS administration is associated with an increase in mitochondrial injury, we monitored effects of AEM-2 on mitochondrial function. AEM-2 significantly reduced mitochondrial superoxide formation, prevented the LPS-induced decrease in mitochondrial membrane potential and attenuated the release of cytochrome c. AEM-2 also inhibited the activities of initiator caspases 8 and 9 and effector caspase 3. The attenuation of apoptosis in AEM-2 treated cells was associated with an increase in cellular autophagy. These data suggest that AEM-2 attenuates cellular injury in LPS-treated THP-1 macrophages and facilitates the removal of cellular debris and damaged organelles via induction of autophagy.

16.
Circ Res ; 97(3): 236-43, 2005 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-16002747

RESUMO

Previous studies suggest that high-density lipoprotein and apoAI inhibit lipopolysaccharide (LPS)-induced inflammatory responses. The goal of the current study was to test the hypothesis that the apoAI mimetic peptide L-4F exerts antiinflammatory effects similar to apoAI. Pretreatment of human umbilical vein endothelial cells (HUVECs) with LPS induced the adhesion of THP-1 monocytes. Incubation of cells with LPS and L-4F (1 to 50 microg/mL) reduced THP-1 adhesion in a concentration-dependent manner. This response was associated with a significant reduction in the synthesis of cytokines, chemokines, and adhesion molecules. L-4F reduced vascular cell adhesion molecule-1 expression induced by LPS or lipid A, whereas a control peptide (Sc-4F) showed no effect. In contrast to LPS treatment, L-4F did not inhibit IL-1beta- or tumor necrosis factor-alpha-induced vascular cell adhesion molecule-1 expression. The inhibitory effect of L-4F on LPS induction of inflammatory markers was associated with reduced binding of LPS to its plasma carrier molecule, lipopolysaccharide binding protein, and decreased binding of LPS to HUVEC monolayers. LPS and L-4F in HUVEC culture medium were fractionated by fast protein liquid chromatography and were localized to the same fractions, suggesting a physical interaction between these molecules. Proinflammatory responses to LPS are associated with the binding of lipid A to cell surface receptors. The current studies demonstrate that L-4F reduces the expression of inflammatory markers induced by LPS and lipid A and suggest that apoAI peptide mimetics may be useful in the treatment of inflammation associated with endotoxemia.


Assuntos
Inflamação/prevenção & controle , Lipopolissacarídeos/antagonistas & inibidores , Peptídeos/farmacologia , Proteínas de Fase Aguda/metabolismo , Sequência de Aminoácidos , Proteínas de Transporte/metabolismo , Adesão Celular , Células Cultivadas , Células Endoteliais/citologia , Humanos , Mediadores da Inflamação/fisiologia , Lipídeo A/farmacologia , Receptores de Lipopolissacarídeos/fisiologia , Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/toxicidade , Lipoproteínas HDL/farmacologia , Glicoproteínas de Membrana/metabolismo , Dados de Sequência Molecular , Monócitos/fisiologia , Peptídeos/metabolismo , Fosfatidilcolinas/farmacologia , Molécula 1 de Adesão de Célula Vascular/biossíntese
17.
Circulation ; 111(23): 3112-8, 2005 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-15939819

RESUMO

BACKGROUND: These studies were designed to determine whether the dual-domain peptide with a class A amphipathic helix linked to the receptor-binding domain of apolipoprotein (apo) E (Ac-hE-18A-NH2) possesses both antidyslipidemic and antiinflammatory properties. METHODS AND RESULTS: A single bolus (15 mg/kg IV) of Ac-hE-18A-NH2 that contains LRKLRKRLLR (141- to 150-residue region of apo E) covalently linked to apo A-I mimetic peptide 18A not only reduced plasma cholesterol levels (baseline, 562+/-29.0 mg/dL versus 287.7+/-22.0 mg/dL at 18 hours, P<0.001) in the Watanabe heritable hyperlipidemic rabbit model but also significantly improved arterial endothelial function. This improvement was associated with a reduction in 2 markers of oxidative stress. First, the plasma lipid hydroperoxide content was reduced significantly, an effect associated with a 5-fold increase in HDL paraoxonase activity. Second, the formation of superoxide anion, a scavenger of nitric oxide, was also significantly reduced in arteries of these animals. CONCLUSIONS: Because dyslipidemia and endothelial dysfunction are common features of the atherosclerotic disease process, this unique dual-domain peptide has ideal composite properties that ameliorate key contributory factors to atherosclerosis.


Assuntos
Apolipoproteínas E/química , Colesterol/sangue , Hiperlipidemias/tratamento farmacológico , Peptídeos/farmacologia , Animais , Apolipoproteínas E/uso terapêutico , Aterosclerose/tratamento farmacológico , Aterosclerose/prevenção & controle , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/patologia , Endotélio Vascular/fisiologia , Hiperlipidemias/patologia , Inflamação/tratamento farmacológico , Erros Inatos do Metabolismo Lipídico/tratamento farmacológico , Lipídeos/sangue , Masculino , Mimetismo Molecular , Estresse Oxidativo/efeitos dos fármacos , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/farmacologia , Fragmentos de Peptídeos/uso terapêutico , Peptídeos/síntese química , Peptídeos/uso terapêutico , Coelhos , Superóxidos/análise
18.
Curr Pharm Biotechnol ; 7(4): 235-40, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16918400

RESUMO

Levels of high density lipoprotein (HDL) and its major protein component, apolipoprotein (apo) A-I, are strongly inversely correlated to risk of atherosclerosis and other vascular diseases. A number of properties of apo A-I may contribute to this protection, including removal of cholesterol from peripheral tissues to the liver (reverse cholesterol transport), anti-inflammatory and anti-oxidative activities, and modulation of vascular function. Apo A-I has lipid-associating domains that form class A amphipathic helices. Peptide analogs that have no sequence homology to the domains in apo A-I but possess the class A motif have been shown to not only associate with phospholipid but also mimic several of the functional properties of apo A-I. Peptide 4F, with four phenylalanines on the non-polar face, was found to be maximally effective in mimicking the positive qualities of apo A-I; this peptide inhibited atherosclerosis, reduced inflammation and oxidation, and improved vascular function in a number of animal models, and when synthesized with D-amino acids is orally bioavailable. Several other classes of peptide mimetics are now being studied, and may contribute to our understanding of the functions of apo E and apo J. The use of peptide mimetics to study apolipoprotein function has proved to be a powerful tool, and may lead to novel therapeutic agents in the prevention of atherosclerosis and other vascular diseases.


Assuntos
Apolipoproteínas/fisiologia , Aterosclerose/tratamento farmacológico , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Doenças Vasculares/tratamento farmacológico , Animais , Apolipoproteína A-I/uso terapêutico , Apolipoproteínas/química , Aterosclerose/fisiopatologia , Colesterol/metabolismo , HDL-Colesterol/metabolismo , Doença das Coronárias/tratamento farmacológico , Humanos , Lipopolissacarídeos/química , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Mimetismo Molecular , Conformação Proteica , Doenças Vasculares/fisiopatologia
19.
Arterioscler Thromb Vasc Biol ; 25(7): 1325-31, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15831812

RESUMO

Despite identical amino acid composition, differences in class A amphipathic helical peptides caused by differences in the order of amino acids on the hydrophobic face results in substantial differences in antiinflammatory properties. One of these peptides is an apolipoprotein A-I (apoA-I) mimetic, D-4F. When given orally to mice and monkeys, D-4F caused the formation of pre-beta high-density lipoprotein (HDL), improved HDL-mediated cholesterol efflux, reduced lipoprotein lipid hydroperoxides, increased paraoxonase activity, and converted HDL from pro-inflammatory to antiinflammatory. In apolipoprotein E (apoE)-null mice, D-4F increased reverse cholesterol transport from macrophages. Oral D-4F reduced atherosclerosis in apoE-null and low-density lipoprotein (LDL) receptor-null mice. In vitro when added to human plasma at nanomolar concentrations, D-4F caused the formation of pre-beta HDL, reduced lipoprotein lipid hydroperoxides, increased paraoxonase activity, and converted HDL from pro-inflammatory to antiinflammatory. Physical-chemical properties and the ability of various class A amphipathic helical peptides to activate lecithin cholesterol acyltransferase (LCAT) in vitro did not predict biologic activity in vivo. In contrast, the use of cultured human artery wall cells in evaluating these peptides was more predictive of their efficacy in vivo. We conclude that the antiinflammatory properties of different class A amphipathic helical peptides depends on subtle differences in the configuration of the hydrophobic face of the peptides, which determines the ability of the peptides to sequester inflammatory lipids. These differences appear to be too subtle to predict efficacy based on physical-chemical properties alone. However, understanding these physical-chemical properties provides an explanation for the mechanism of action of the active peptides.


Assuntos
Apolipoproteína A-I/química , Apolipoproteína A-I/metabolismo , Mimetismo Molecular , Peptídeos/química , Peptídeos/metabolismo , Animais , Humanos , Estrutura Secundária de Proteína
20.
Circulation ; 109(25): 3215-20, 2004 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-15197147

RESUMO

BACKGROUND: These studies were designed to determine the mechanism of action of an oral apolipoprotein (apo) A-I mimetic peptide, D-4F, which previously was shown to dramatically reduce atherosclerosis in mice. METHODS AND RESULTS: Twenty minutes after 500 microg of D-4F was given orally to apoE-null mice, small cholesterol-containing particles (CCPs) of 7 to 8 nm with pre-beta mobility and enriched in apoA-I and paraoxonase activity were found in plasma. Before D-4F, both mature HDL and the fast protein liquid chromatography fractions containing the CCPs were proinflammatory. Twenty minutes after oral D-4F, HDL and CCPs became antiinflammatory, and there was an increase in HDL-mediated cholesterol efflux from macrophages in vitro. Oral D-4F also promoted reverse cholesterol transport from intraperitoneally injected cholesterol-loaded macrophages in vivo. In addition, oral D-4F significantly reduced lipoprotein lipid hydroperoxides (LOOH), except for pre-beta HDL fractions, in which LOOH increased. CONCLUSIONS: The mechanism of action of oral D-4F in apoE-null mice involves rapid formation of CCPs, with pre-beta mobility enriched in apoA-I and paraoxonase activity. As a result, lipoprotein LOOH are reduced, HDL becomes antiinflammatory, and HDL-mediated cholesterol efflux and reverse cholesterol transport from macrophages are stimulated.


Assuntos
Apolipoproteína A-I/farmacologia , Apolipoproteínas E/deficiência , Arteriosclerose/genética , Colesterol/metabolismo , Hiperlipoproteinemia Tipo II/genética , Lipoproteínas HDL/biossíntese , Macrófagos Peritoneais/efeitos dos fármacos , Administração Oral , Sequência de Aminoácidos , Animais , Apolipoproteína A-I/uso terapêutico , Apolipoproteínas E/genética , Arteriosclerose/sangue , Arildialquilfosfatase/sangue , Transporte Biológico/efeitos dos fármacos , Células Cultivadas , Quimiotaxia/efeitos dos fármacos , Técnicas de Cocultura , Avaliação Pré-Clínica de Medicamentos , Feminino , Lipoproteínas de Alta Densidade Pré-beta , Humanos , Hiperlipoproteinemia Tipo II/sangue , Inflamação , Peroxidação de Lipídeos/efeitos dos fármacos , Lipoproteínas HDL/sangue , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Monócitos/citologia , Monócitos/efeitos dos fármacos , Monócitos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA