Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Biosci Biotechnol Biochem ; 80(7): 1379-81, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26932266

RESUMO

In this study, we investigated the effect of TGF-ß1 on cholesterol synthesis in human keratinocytes. TGF-ß1 increased the level of cholesterol and the mRNA level of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase in human keratinocytes. These results show that TGF-ß1 induces cholesterol synthesis by increasing HMG-CoA reductase mRNA expression in human keratinocytes.


Assuntos
Acil Coenzima A/biossíntese , Colesterol/biossíntese , Hidroximetilglutaril-CoA Redutases/genética , RNA Mensageiro/genética , Fator de Crescimento Transformador beta1/farmacologia , Linhagem Celular , Colesterol/agonistas , Expressão Gênica , Humanos , Hidroximetilglutaril-CoA Redutases/metabolismo , Queratinócitos/citologia , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , RNA Mensageiro/agonistas , RNA Mensageiro/metabolismo
2.
Arch Toxicol ; 90(12): 3045-3060, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26821219

RESUMO

The piperazine derivatives most frequently consumed for recreational purposes are 1-benzylpiperazine, 1-(3,4-methylenedioxybenzyl) piperazine, 1-(3-trifluoromethylphenyl) piperazine and 1-(4-methoxyphenyl) piperazine. Generally, they are consumed as capsules, tablets or pills but also in powder or liquid forms. Currently, the precise mechanism by which piperazine designer drugs induce hepatotoxicity and whether they act by a common pathway is unclear. To answer this question, we performed a gene array study with rat hepatocytes incubated with the four designer drugs. Non-cytotoxic concentrations were chosen that neither induce a decrease in reduced glutathione or ATP depletion. Analysis of the gene array data showed a large overlap of gene expression alterations induced by the four drugs. This 'piperazine designer drug consensus signature' included 101 up-regulated and 309 down-regulated probe sets (p < 0.05; FDR adjusted). In the up-regulated genes, GO groups of cholesterol biosynthesis represented a dominant overrepresented motif. Key enzymes of cholesterol biosynthesis up-regulated by all four piperazine drugs include sterol C4-methyloxidase, isopentyl-diphosphate-Δ-isomerase, Cyp51A1, squalene epoxidase and farnesyl diphosphate synthase. Additionally, glycoprotein transmembrane nmb, which participates in cell adhesion processes, and fatty acid desaturase 1, an enzyme that regulates unsaturation of fatty acids, were also up-regulated by the four piperazine designer drugs. Regarding the down-regulated probe sets, only one gene was common to all four piperazine derivatives, the betaine-homocysteine-S-methyltransferase 2. Analysis of transcription factor binding sites of the 'piperazine designer drug consensus signature' identified the sterol regulatory element binding protein (SREBP-1) as strongly overrepresented in the up-regulated genes. SREBP transcription factors are known to regulate multiple genes of cholesterol metabolism. In conclusion, the present study shows that piperazine designer drugs act by up-regulating key enzymes of cholesterol biosynthesis which is likely to increase the risk of phospholipidosis and steatosis.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/enzimologia , Colesterol/agonistas , Drogas Desenhadas/toxicidade , Indução Enzimática/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Metabolismo dos Lipídeos/efeitos dos fármacos , Piperazinas/toxicidade , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Colesterol/biossíntese , Perfilação da Expressão Gênica , Hepatócitos/metabolismo , Hepatócitos/patologia , Concentração Inibidora 50 , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Concentração Osmolar , Análise de Componente Principal , Ratos Wistar
3.
Lipids Health Dis ; 14: 130, 2015 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-26493087

RESUMO

BACKGROUND: Sphingomyelin (SM) and cholesterol are two types of lipid closely related biophysically. Treating the cells with exogenous sphingomyelinase (SMase) induces trafficking of cholesterol from membrane to intracellular pools and inhibition of cholesterol synthesis. In the present work, we address a question whether increased cholesterol synthesis affects hydrolysis of SM by endogenous SMases. METHODS: Both HepG2 and Caco-2 cells were incubated with mevalonate. The SMase activity was determined and its mRNA examined by qPCR. The cellular levels of cholesterol, SM, and phosphatidylcholine (PC) were determined and cell proliferation rate assayed. RESULTS: We found that mevalonate dose-dependently decreased acid but not neutral SMase activity in both HepG2 and Caco-2 cells with HepG2 cells being more sensitive to mevalonate. Kinetic examination in HepG2 cells revealed that acid SMase activity was increasing with cell proliferation, and such an increase was reversed by mevalonate treatment. Acid SMase mRNA was not significantly decreased and Western blot showed signs of proteolysis of acid SMase by mevalonate. After mevalonate treatment, the levels of cholesterol were significantly increased associated with increases in SM and PC. The cell growth was retarded by mevalonate and the effect was more obvious in HepG2 cells than in Caco-2 cells. CONCLUSION: Mevalonate can trigger a mechanism to enhance SM levels by inhibition of acid SMase. The effect may ensure the coordinate changes of SM and cholesterol in the cells. Mevalonate also affects cell growth with mechanism required further characterization.


Assuntos
Antineoplásicos/farmacologia , Colesterol/agonistas , Ácido Mevalônico/farmacologia , RNA Mensageiro/antagonistas & inibidores , Esfingomielina Fosfodiesterase/antagonistas & inibidores , Esfingomielinas/agonistas , Células CACO-2 , Proliferação de Células/efeitos dos fármacos , Colesterol/biossíntese , Relação Dose-Resposta a Droga , Expressão Gênica , Células Hep G2 , Humanos , Hidrólise , Cinética , Especificidade de Órgãos , Fosfatidilcolinas/agonistas , Fosfatidilcolinas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Esfingomielina Fosfodiesterase/genética , Esfingomielina Fosfodiesterase/metabolismo , Esfingomielinas/metabolismo
4.
Pharm Biol ; 53(10): 1481-7, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25857322

RESUMO

CONTEXT: Chrysin, a natural flavonoid, has been shown to possess multiple pharmacological activities including anti-atherosclerosis. OBJECTIVE: The effects of chrysin on foam cell formation and cholesterol flow in RAW264.7 macrophages were investigated in this work to explore the potential mechanism underlying its anti-atherogenic activity. MATERIALS AND METHODS: The inhibitive effect of chrysin on foam cell formation and cholesterol accumulation induced by oxidized low-density lipoprotein cholesterol (ox-LDL) was assessed by oil red O staining and intracellular total cholesterol and triglyceride quantification in RAW264.7 macrophages. The action of chrysin on cholesterol efflux and influx was tested by fluorescent assays. Real-time quantitative PCR was used to quantify the relative expression of cholesterol flow-associated genes and luciferase assay was applied to test the transcription activity of peroxisome proliferator-activated receptor gamma (PPARγ). RESULTS: Chrysin dose dependently inhibited the formation of foam cells and prevented the enhanced cholesterol accumulation by ox-LDL. Treatment with chrysin (10 µM) significantly enhanced cholesterol efflux and substantially inhibited cholesterol influx. Simultaneously, chrysin significantly increased the mRNA levels of PPARγ, liver X receptor alpha (LXRα), ATP-binding cassette, sub-family A1 (ABCA1), and sub-family G1 (ABCG1), decreased scavenger receptor A1 (SR-A1) and SR-A2, and increased the transcriptional activity of PPARγ. DISCUSSION AND CONCLUSION: Chrysin is a new inhibitor of foam cell formation that may stimulate cholesterol flow. Up-regulation of the classical PPARγ-LXRα-ABCA1/ABCG1 pathway and down-regulation of SR-A1 and SR-A2 may participate in its suppressive effect on intracellular cholesterol accumulation.


Assuntos
Colesterol/metabolismo , Flavonoides/farmacologia , Células Espumosas/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Pandanaceae , Animais , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Linhagem Celular , Colesterol/agonistas , Relação Dose-Resposta a Droga , Flavonoides/isolamento & purificação , Células Espumosas/metabolismo , Macrófagos/metabolismo , Camundongos
5.
J Lipid Res ; 51(1): 202-9, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19578163

RESUMO

Cholesterol homeostasis, defined as the balance between absorption and synthesis, influences circulating cholesterol concentrations and subsequent coronary heart disease (CHD) risk. Statin therapy targets the rate-limiting enzyme in cholesterol biosynthesis and is efficacious in lowering CHD events and mortality. Nonetheless, CHD events still occur in some treated patients. To address differences in outcome during pravastatin therapy (40 mg/day), plasma markers of cholesterol synthesis (desmosterol, lathosterol) and fractional cholesterol absorption (campesterol, sitosterol) were measured, baseline and on treatment, in the Prospective Study of Pravastatin in the Elderly at Risk trial participants with (cases, n = 223) and without (controls, n = 257) a CHD event. Pravastatin therapy decreased plasma LDL-cholesterol and triglycerides and increased HDL-cholesterol concentrations to a similar extent in cases and controls. Decreased concentrations of the cholesterol synthesis markers desmosterol (-12% and -11%) and lathosterol (-50% and -56%) and increased concentrations of the cholesterol absorption markers campesterol (48% and 51%) and sitosterol (25% and 26%) were observed on treatment, but the magnitude of change was similar between cases and controls. These data suggest that decreases in cholesterol synthesis in response to pravastatin treatment were accompanied by modest compensatory increases in fractional cholesterol absorption. The magnitude of these alterations were similar between cases and controls and do not explain differences in outcomes with pravastatin treatment.


Assuntos
Doença das Coronárias/tratamento farmacológico , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Pravastatina/uso terapêutico , Idoso , Idoso de 80 Anos ou mais , Colesterol/agonistas , Colesterol/análogos & derivados , Colesterol/sangue , HDL-Colesterol/agonistas , HDL-Colesterol/sangue , LDL-Colesterol/antagonistas & inibidores , LDL-Colesterol/sangue , Doença das Coronárias/sangue , Doença das Coronárias/mortalidade , Desmosterol/antagonistas & inibidores , Desmosterol/sangue , Feminino , Homeostase/efeitos dos fármacos , Homeostase/fisiologia , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/administração & dosagem , Masculino , Fitosteróis/agonistas , Fitosteróis/sangue , Pravastatina/administração & dosagem , Sitosteroides/agonistas , Sitosteroides/sangue , Triglicerídeos/antagonistas & inibidores , Triglicerídeos/sangue
6.
Am J Clin Nutr ; 102(6): 1339-46, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26561632

RESUMO

BACKGROUND: Adverse effects of industrially produced trans fatty acids (iTFAs) on the risk of coronary artery disease are well documented in the scientific literature; however, effects of naturally occurring trans fatty acids (TFAs) from ruminant animals (rTFA), such as vaccenic acid (VA) and cis-9,trans-11 conjugated linoleic acid (c9,t11-CLA), are less clear. Although animal and cell studies suggest that VA and c9,t11-CLA may be hypocholesterolemic and antiatherogenic, epidemiologic data comparing rTFAs and iTFAs are inconsistent, and human intervention studies have been limited, underpowered, and not well controlled. OBJECTIVE: We determined the effects of VA, c9,t11-CLA, and iTFA, in the context of highly controlled diets (24 d each), on lipoprotein risk factors compared with a control diet. RESULTS: We conducted a double-blind, randomized, crossover feeding trial in 106 healthy adults [mean ± SD age: 47 ± 10.8 y; body mass index (in kg/m(2)): 28.5 ± 4.0; low-density lipoprotein (LDL) cholesterol: 3.24 ± 0.63 mmol/L]. Diets were designed to have stearic acid replaced with the following TFA isomers (percentage of energy): 0.1% mixed isomers of TFA (control), ∼3% VA, ∼3% iTFA, or 1% c9,t11-CLA. Total dietary fat (34% of energy) and other macronutrients were matched. Total cholesterol (TC), LDL cholesterol, triacylglycerol, lipoprotein(a), and apolipoprotein B were higher after VA than after iTFA; high-density lipoprotein (HDL) cholesterol and apolipoprotein AI also were higher after VA. Compared with control, VA and iTFA both increased TC, LDL cholesterol, ratio of TC to HDL cholesterol, and apolipoprotein B (2-6% change; P < 0.05); VA also increased HDL cholesterol, apolipoprotein AI, apolipoprotein B, and lipoprotein(a) (2-6% change; P < 0.05), whereas iTFA did not. c9,t11-CLA lowered triacylglycerol (P ≤ 0.01) and had no effect on other lipoprotein risk factors. CONCLUSIONS: With respect to risk of cardiovascular disease, these results are consistent with current nutrition labeling guidelines, with the requirement of VA, but not c9,t11-CLA, to be listed under TFA on the Nutrition Facts Panel. This trial was registered at clinicaltrials.gov as NCT00942656.


Assuntos
LDL-Colesterol/agonistas , Gorduras Insaturadas na Dieta/efeitos adversos , Hipercolesterolemia/etiologia , Ácidos Linoleicos Conjugados/efeitos adversos , Ácidos Oleicos/efeitos adversos , Óleos de Plantas/efeitos adversos , Ácidos Graxos trans/efeitos adversos , Adulto , Doenças Cardiovasculares/epidemiologia , Doenças Cardiovasculares/etiologia , Colesterol/agonistas , Colesterol/sangue , LDL-Colesterol/sangue , Estudos Cross-Over , Método Duplo-Cego , Feminino , Humanos , Hidrogenação , Hipercolesterolemia/sangue , Hipercolesterolemia/fisiopatologia , Hipertrigliceridemia/sangue , Hipertrigliceridemia/etiologia , Hipertrigliceridemia/fisiopatologia , Masculino , Pessoa de Meia-Idade , Óleos de Plantas/química , Fatores de Risco , Triglicerídeos/agonistas , Triglicerídeos/sangue
7.
Biofactors ; 41(4): 273-88, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26228307

RESUMO

Atherogenesis is associated with macrophage cholesterol and oxidized lipids accumulation and foam cell formation. However, two other major lipid-metabolizing cell classes, namely intestinal and liver cells, are also associated with atherogenesis. This study demonstrates that manipulations of cellular oxidative stress (by fatty acids, glucose, low-density lipoprotein, angiotensin II, polyphenolic antioxidants, or the glutathione/paraoxonase 1 systems) have some similar, but also some different effects on cholesterol metabolism in macrophages (J774A.1) versus intestinal cells (HT-29) versus liver cells (HuH7). Cellular oxidative stress was ≈3.5-folds higher in both intestinal and liver cells versus macrophages. In intestinal cells or liver cells versus macrophages, the cholesterol biosynthesis rate was increased by 9- or 15-fold, respectively. In both macrophages and intestinal cells C-18:1 and C-18:2 but not C-18:0, fatty acids significantly increased oxidative stress, whereas in liver cells oxidative stress was significantly decreased by all three fatty acids. In liver cells, trans C-18:1 versus cis C-18:1, unlike intestinal cells or macrophages, significantly increased cellular oxidative stress and cellular cholesterol biosynthesis rate. Pomegranate juice (PJ), red wine, or their phenolics gallic acids or quercetin significantly reduced cellular oxidation mostly in macrophages. Recombinant PON1 significantly decreased macrophage (but not the other cells) oxidative stress by ≈30%. We conclude that cellular atherogenesis research should look at atherogenicity, not only in macrophages but also in intestinal and liver cells, to advance our understanding of the complicated mechanisms behind atherogenesis. © 2015 BioFactors, 41(4):273-288, 2015.


Assuntos
Antioxidantes/farmacologia , Colesterol/biossíntese , Células Epiteliais/metabolismo , Hepatócitos/metabolismo , Macrófagos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Angiotensina II/farmacologia , Animais , Antioxidantes/isolamento & purificação , Arildialquilfosfatase/farmacologia , Linhagem Celular Tumoral , Colesterol/agonistas , Cumarínicos/isolamento & purificação , Cumarínicos/farmacologia , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Ácidos Graxos/farmacologia , Ácidos Graxos Insaturados/farmacologia , Frutas/química , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Lipoproteínas LDL/farmacologia , Lythraceae/química , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Camundongos , Especificidade de Órgãos , Estresse Oxidativo/efeitos dos fármacos , Fenóis/isolamento & purificação , Fenóis/farmacologia , Espécies Reativas de Oxigênio/agonistas , Espécies Reativas de Oxigênio/antagonistas & inibidores , Proteínas Recombinantes/farmacologia , Ácidos Graxos trans/farmacologia , Vinho/análise
8.
Atherosclerosis ; 146(2): 249-57, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10532681

RESUMO

Angiotensin II (Ang II) was shown to be an important risk factor for accelerated atherosclerosis. Inhibition of Ang II action on the arterial wall by blocking its production with angiotensin converting enzyme (ACE) inhibitors, or by blocking binding to its receptors on cells with antagonists was shown to attenuate atherogenesis in animal model of atherosclerosis. We questioned whether Ang II atherogenicity is related to a stimulatory effect of Ang II on macrophage cholesterol biosynthesis. Angiotensin II injected intraperitoneally once a day (0.1 ml of 10(-7) M per mouse) for a period of 30 days, to the apolipoprotein E deficient mice increased the atherosclerotic lesion area by 95% (P < 0.01 vs. control), compared to placebo-injected mice, with no significant effect on blood pressure or on plasma cholesterol levels. On using mouse peritoneal macrophages (MPMs) that were harvested after intraperitoneally injection of Ang II, an increased rate of cellular cholesterol biosynthesis (measured as incorporation of [3H]acetate into cholesterol) by up to 90% (P < 0.01 vs. control) was observed. In mice treated with the ACE inhibitor, Fosinopril (25 mg/kg per day) a reduction in their MPM's cholesterol synthesis by up to 70% (P < 0.01 vs. control) was obtained. In vitro studies in human monocyte-derived macrophages (HMDM), in MPMs from control BALB/c mice, and in J-774 A.1 macrophage-like cell line demonstrated up to 44, 34 and 30% stimulation of macrophage cholesterol biosynthesis, respectively, following cell incubation with 10(-7) M Ang II for 18 h at 37 degrees C. The stimulatory effect of Ang II on macrophage cholesterol biosynthesis could be related to its interaction with the macrophage AT1 receptor, as Losartan (10(-5) M), an AT1 blocker, but not PD 123319 (10(-5) M), an AT2 blocker, prevented the stimulatory effect on macrophage cholesterol synthesis. Furthermore, in cells that lack the AT1 receptor (RAW macrophages), Ang II did not increase cellular cholesterol synthesis. Ang II increased macrophage 3-hydroxy-3-methyl glutaryl CoA (HMG CoA) reductase mRNA levels in a dose dependent manner in J-774 A.1 macrophages and in MPM. Losartan, the AT1 receptor antagonist clearly attenuated this mRNA induction. We thus conclude that Ang II stimulation of macrophage cholesterol biosynthesis is related to its interaction with the AT1 receptor, followed by stimulation of macrophage HMG CoA reductase gene expression, which leads to increased cellular cholesterol biosynthesis, and can possibly result in macrophage cholesterol accumulation and foam cell formation.


Assuntos
Angiotensina II/farmacologia , Apolipoproteínas E/deficiência , Arteriosclerose/etiologia , Colesterol/biossíntese , Macrófagos Peritoneais/metabolismo , Monócitos/metabolismo , Vasoconstritores/farmacologia , Antagonistas de Receptores de Angiotensina , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Animais , Arteriosclerose/metabolismo , Arteriosclerose/patologia , Linhagem Celular , Colesterol/agonistas , Colesterol/metabolismo , Primers do DNA/química , Modelos Animais de Doenças , Expressão Gênica/efeitos dos fármacos , Humanos , Hidroximetilglutaril-CoA Redutases/biossíntese , Hidroximetilglutaril-CoA Redutases/genética , Injeções Intraperitoneais , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/patologia , Camundongos , Monócitos/efeitos dos fármacos , Monócitos/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor Tipo 1 de Angiotensina , Receptor Tipo 2 de Angiotensina , Receptores de Angiotensina/agonistas , Receptores de Angiotensina/genética , Receptores de Angiotensina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Risco
9.
J Lipid Res ; 42(7): 1125-33, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11441141

RESUMO

Endogenous expression of apolipoprotein (apo)E in macrophages facilitates cholesterol efflux in the presence and absence of extracellular sterol acceptors. A proteoglycan-associated pool of apoE has also been described. The relationship between a proteoglycan-associated pool of apoE and enhanced cholesterol efflux was investigated in these studies. Inhibition of proteoglycan expression reduced cholesterol efflux from apoE-expressing cells ( J774E(+)) in the presence and absence of HDL, but did not do so from nonexpressing cells ( J774E(-)). The effect of proteoglycan depletion on sterol efflux from J774E(+) cells was confirmed by measuring differences in cell sterol mass, secreted sterol mass, and sterol efflux rates. Furthermore, apoE-containing particles secreted from proteoglycan-depleted J774E(+) cells were denser than those secreted from J774E(+) cells with intact proteoglycan expression. Also, in J774E(+) cells with intact proteoglycans, apoE particles isolated from the cell surface proteoglycan layer were denser than secreted particles. The apoE-lipid particles isolated from the cell surface proteoglycan layer had a lower lipid-to-apoE and cholesterol-to-apoE ratio compared with secreted particles. In distinction, proteoglycan depletion of J774E(-) cells did not reduce sterol efflux produced by the exogenous addition of apoE. These observations indicate that one mechanism by which endogenous expression of apoE facilitates effective cholesterol efflux from macrophages is related to its retention at the cell surface in a proteoglycan-associated pool. Further, our data suggest that apoE arrives at the cell surface in a relatively lipid-poor state, and that a proximate source of lipid available to the proteoglycan-bound apoE at the cell surface resides in the plasma membrane.


Assuntos
Apolipoproteínas E/biossíntese , Macrófagos/metabolismo , Proteoglicanas/metabolismo , Animais , Sítios de Ligação/fisiologia , Linhagem Celular/citologia , Colesterol/agonistas , Colesterol/metabolismo , Heparina Liase/farmacologia , Lipoproteínas HDL/metabolismo , Lipoproteínas HDL3 , Macrófagos/citologia , Proteoglicanas/química , Proteoglicanas/efeitos dos fármacos
10.
São Paulo; s.n; s.n; mai. 2013. 181 p. tab, graf, ilus.
Tese em Português | LILACS | ID: biblio-837036

RESUMO

O processo inflamatório tem um papel fundamental na gênese e desenvolvimento da aterosclerose, sendo que a disfunção endotelial é considerada um dos estágios iniciais da aterogênese. Por meio da inibição da enzima hidroxi-metil-glutaril coA redutase (HMGCR), as estatinas reduzem a biossíntese do colesterol e a formação de isoprenóides, produtos intermediários da síntese do colesterol que são importantes na modificação pós-transcricional de GTPases pequenas que estão envolvidas na disfunção endotelial e inflamação vascular. A ezetimiba é um inibidor da absorção do colesterol através da inibição da proteína NPC1L1. Com a finalidade de esclarecer os mecanismos moleculares da inibição da síntese e da absorção do colesterol sobre a modulação de biomarcadores inflamatórios e de adesão celular foram utilizados modelos in vitro com células endoteliais (HUVEC) e monócitos (células THP-1), e in vivo com células mononucleares do sangue periférico (CMSP) de indivíduos hipercolesterolêmicos (HC). O efeito das estatinas, atorvastatina e sinvastatina, e da ezetimiba na expressão de RNAm e proteínas de moléculas de adesão endoteliais e moduladores do processo inflamatório, como citocinas e óxido nítrico (NO), foi estudado em células HUVEC. O efeito desses fármacos sobre a expressão de moléculas de adesão monocitárias foi estudado em células THP-1. O efeito da terapia hipolipemiante sobre essas moléculas foi também estudada em CMSP de HC tratados com ezetimiba (10 mg/dia/4 semanas), sinvastatina (10 mg/dia/8 semanas) e sinvastatina combinada com ezetimiba (10 mg de cada/dia/4 semanas). A expressão de RNAm foi avaliada por RT-qPCR. A expressão de moléculas de adesão na superfície de células THP-1 e HUVEC foi estudada por citometria de fluxo. A quantificação de citocinas secretadas no sobrenadante de células HUVEC e no plasma dos HC foi analisada pela tecnologia Milliplex. A quantificação do perfil lipífico, Proteína C reativa ultra-sensível (PCRus) e NO foi realizada por métodos laboratoriais convencionais. O papel do NO na modulação dos marcadores inflamatórios pelas estatinas foi também estudada, usando modelo de células HUVEC com NOS3 silenciado por interferência de RNAm e também por meio do uso do inibidor da síntese do óxido nítrico, L-NAME. Também foi avaliado o efeito de hipolipemiantes na expressão dos microRNAs (miRs) 221, miR-222 e miR-1303 em células HUVEC por meio do stem-loop RT-qPCR. O tratamento com atorvastatina e sinvastatina reduziu a expressão de RNAm e proteínas das moléculas de adesão LSelectina, PSGL-1 e VLA-4, em células THP-1 pré-tratadas com TNFα por 12 h. A ezetimiba reduziu a expressão de L-Selectina apenas no nível transcricional. Em células HUVEC, as estatinas diminuíram a expressão de RNAm de IL1B e SELP, entretanto aumentaram a de VCAM1. A ezetimiba reduziu a expressão de RNAm do IL1B. Entretanto as expressões de SELE, MMP9, IL6 e MMP9 não foram afetadas pelos tratamentos. A expressão das proteínas ICAM-1 e P-Selectina, na superfície de células HUVEC, foi diminuída pelo tratamento com as estatinas, mas não pela ezetimiba. Da mesma forma, a secreção das citocinas IL-6 e MCP-1 foram reduzidas pelas estatinas, entretanto a secreção de IL-8 não foi modificada por nenhum dos tratamentos. A expressão de NOS3 e a liberação de NO em células HUVEC foi aumentada pelas estatinas, porém não foi estimulada pela ezetimiba. Entretanto, os efeitos antiinflamatórios exercidos pelas estatinas foram independentes dessa via devido a que estes efeitos foram mantidos em células HUVEC com NOS3 silenciado por interferência de RNAm. Apesar de que o efeito sobre ICAM-1 e MCP-1 foi atenuado quando as células foram simultaneamente tratadas com L-NAME, os efeitos das estatinas parecem ser independentes da liberação de NO. As estatinas e a ezetimiba reduziram a expressão do miR-221, em células HUVEC. A expressão do miR-222 foi reduzida só pelo tratamento com atorvastatina. A expressão do miR-1303 não foi modulada pelos tratamentos hipolipemiantes. Em pacientes HC, a terapia de associação da sinvastatina e ezetimiba demonstrou melhorar o perfil lipídico de forma mais efetiva que ambas monoterapias. Da mesma forma, o tratamento combinado resultou em maior beneficio pela redução da expressão de RNAm em CMSP e da concentração plasmática das proteínas IL-1 ß, MCP-1, IL-8 e TNFα. A expressão de ICAM1 foi diminuída apenas no nível transcricional, entretanto a expressão de RNAm mas não da proteína do TNFα foi também reduzida pela sinvastatina em monoterapia. Não houve modulação de RNAm ou proteínas de outros marcadores estudados no modelo in vivo. Por outro lado, os efeitos anti-inflamatórios observados nos indivíduos HC foram independentes da modulação de PCRus e NO que não foram modificados pelos tratamentos hipolipemiantes. Neste estudo, foram confirmados os propostos efeitos pleiotrópicos das estatinas em modelos células de monócito e endotélio vascular in vitro e em pacientes HC. Por outro lado, apesar de ser menos potente que as estatinas foi mostrado que a inibição da absorção do colesterol tem também um efeito anti-inflamatório. A redução adicional do colesterol causado pela combinação das terapias hipolipemiantes outorga um maior beneficio cardiovascular em pacientes hipercolesterolêmicos


The inflammatory process has a key role in the genesis and development of atherosclerosis and the endothelial disfunction is considered as a first step in atherogenesis. By inhibiting the hydroxyl-methyl-glutaryl coA reductase (HMGCR)m statins reduce the cholesterol synthesis and isoprenoid generation, which are intermediary products of cholesterol synthesis with important role in posttranscriptional modifications of small GTPases that are involved in endothelial disfunction and vascular inflammation. The ezetimibe is an inhibitor of cholesterol absorption by inhibiting the NPC1L1 protein. To clarify the molecular mechanisms of the inhibition of cholesterol synthesis and absorption modulating inflammatory and cell adhesion biomarkers we used in vitro models of endothelial cells (HUVEC) and monocytes (THP-1), and an in vivo model of peripheral blood mononuclear cells (PBMC) from hypercholesterolemic (HC) patients. The effect of the statins, atorvastatin and simvastatin, and the ezetimibe on mRNA and protein expression of endothelial adhesion molecules and modulators of the inflammatory process, as citokynes and nitric oxide (NO), was analyzed in HUVEC. The effect of these drugs on the expression of monocyte adhesion molecules was also studied in THP-1. The influence of hypolipemiant therapy on the adhesion molecules was also analyzed in PBMC from HC treated with ezetimibe (10 mg/day/4-weeks), simvastatin (10 mg/day/8-weeks) and simvastatin combined with ezetimibe (10 mg each/day/4-weeks). The mRNA expression was evaluated by RT-qPCR. The expression of adhesion molecules on the surface of THP-1 and HUVEC cells was analyzed flow cytometry. The citokynes in the supernatants of HUVEC were quantified using the milliplex technology. The Lipid profile, high-sensivity PCR (hsPCR) and NO were determined by conventional laboratory methods. The role of the NO on the statin-modulation of inflammatory markers was also studied using a model with silenced NOS3 by interference of mRNA and by the use of the inhibitor of NO synthesis, L-NAME. The effect of hypolipemiants on the expression of microRNAs (miRs) 221, miR-222 and miR-1303 was also evaluated in HUVEC using the stem-loom RT-qPCR. Atorvastatin and simvastatin reduced the mRNA and protein expression of the adhesion molecules L-Selectin, PSGL-1 and VLA-4 in THP-1 cells pre-treated with TNFα for 12 h. The ezetimibe reduced the L-Selectin expression only at transcriptional level. In HUVEC, statins diminished IL1B and SELP mRNA expression, whereas VCAM1 was increased. The ezetimibe reduced the IL1B mRNA expression. However, SELE, MMP9, IL6 and MMP9 mRNA expressions were not affected by the treatments. The protein expression of ICAM-1 and P-Selectin on the surface of HUVEC was reduced by statins, but not by the ezetimibe. Similarly, IL-6 and MCP-1 secretion were reduced by statins, whereas IL-8 secretion was not modified by the treatments. The NO release and NOS3 expression in HUVEC was increased by the statins, however it was not stimulated by ezetimibe. Moreover, the anti-inflammatory statin effects were independent of this pathway due to statin effects were maintained in HUVEC with silenced NOS3. Although the statin effect on ICAM-1 and MCP-1 were attenuated by L-NAME co treatment, the statin effects seem to be independent of NO release. Statins and ezetimibe reduced miR221 in HUVEC. miR-222 expression was reduced only by atorvastatin. miR-1303 was not affected by the treatments. In HC patients, the improvement of the lipid profile simvastatin combined with ezetimibe was more efficient than both monotherapies. Similarly, the association therapy was better in reducing the mRNA expression in PBMC and plasma concentration of IL-1ß, MCP-1, IL-8 and TNFα. ICAM1 expression was reduced only at transcriptional level, whereas mRNA but not protein expression of TNFα was also reduced by the simvastatin monotherapy. There was no modulation mRNA or protein expression of other studied markers in the in vivo model. Additionally, the anti-inflammatory effects observed in the HC were independent of PCRus or NO modulation, which were not altered by the hypolipemiant treatments. In this study, the proposed plitropic effects of statins were confirmed in monocytes and endothelial cells in vitro and in HC patients. Moreover, although it was less potent than statins, an anti-inflammatory effect was also observed for the inhibition of cholesterol absorption. An additional reduction of the cholesterol caused by combined hypolipemiant therapies gives a greater cardiovascular beneffict in hypercholesterolemic patients


Assuntos
Biomarcadores/metabolismo , Colesterol/agonistas , Inibidores Enzimáticos/farmacologia , Absorção Fisiológica , Inflamação/classificação , Moléculas de Adesão Celular , Adesão Celular , Inibidores de Hidroximetilglutaril-CoA Redutases/análise , Ezetimiba/administração & dosagem , Hipercolesterolemia/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA