Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Front Endocrinol (Lausanne) ; 12: 601160, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33815271

RESUMEN

Background: Non-alcoholic fatty liver disease (NAFLD) is a complex multifactorial disorder that is characterised by dysfunctional lipid metabolism and cholesterol homeostasis, and a related chronic inflammatory response. NAFLD has become the most common cause of chronic liver disease in many countries, and its prevalence continues to rise in parallel with increasing rates of obesity. Here, we evaluated the putative NAFLD-attenuating effects of a multicomponent medicine consisting of 24 natural ingredients: Hepar compositum (HC-24). Methods: Ldlr-/-.Leiden mice were fed a high-fat diet (HFD) with a macronutrient composition and cholesterol content comparable to human diets for 24 weeks to induce obesity-associated metabolic dysfunction, including hepatic steatosis and inflammation. HC-24 or vehicle control was administered intraperitoneally 3 times/week (1.5 ml/kg) for the last 18 weeks of the study. Histological analyses of liver and adipose tissue were combined with extensive hepatic transcriptomics analysis. Transcriptomics results were further substantiated with ELISA, immunohistochemical and liver lipid analyses. Results: HFD feeding induced obesity and metabolic dysfunction including adipose tissue inflammation and increased gut permeability. In the liver, HFD-feeding resulted in a disturbance of cholesterol homeostasis and an associated inflammatory response. HC-24 did not affect body weight, metabolic risk factors, adipose tissue inflammation or gut permeability. While HC-24 did not alter total liver steatosis, there was a pronounced reduction in lobular inflammation in HC-24-treated animals, which was associated with modulation of genes and proteins involved in inflammation (e.g., neutrophil chemokine Cxcl1) and cholesterol homeostasis (i.e., predicted effect on 'cholesterol' as an upstream regulator, based on gene expression changes associated with cholesterol handling). These effects were confirmed by CXCL1 ELISA, immunohistochemical staining of neutrophils and biochemical analysis of hepatic free cholesterol content. Intrahepatic free cholesterol levels were found to correlate significantly with the number of inflammatory aggregates in the liver, thereby providing a potential rationale for the observed anti-inflammatory effects of HC-24. Conclusions: Free cholesterol accumulates in the liver of Ldlr-/-.Leiden mice under physiologically translational dietary conditions, and this is associated with the development of hepatic inflammation. The multicomponent medicine HC-24 reduces accumulation of free cholesterol and has molecular and cellular anti-inflammatory effects in the liver.


Asunto(s)
Colesterol/metabolismo , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Extractos Vegetales/administración & dosificación , Animales , Quimiocina CXCL1/genética , Quimiocina CXCL1/metabolismo , Dieta Alta en Grasa/efectos adversos , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/inmunología , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/inmunología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Receptores de LDL/genética , Receptores de LDL/inmunología
2.
Int J Mol Sci ; 20(18)2019 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-31491949

RESUMEN

Obesity characterized by adiposity and ectopic fat accumulation is associated with the development of non-alcoholic fatty liver disease (NAFLD). Treatments that stimulate lipid utilization may prevent the development of obesity and comorbidities. This study evaluated the potential anti-obesogenic hepatoprotective effects of combined treatment with L-carnitine and nicotinamide riboside, i.e., components that can enhance fatty acid transfer across the inner mitochondrial membrane and increase nicotinamide adenine nucleotide (NAD+) levels, which are necessary for ß-oxidation and the TCA cycle, respectively. Ldlr -/-.Leiden mice were treated with high-fat diet (HFD) supplemented with L-carnitine (LC; 0.4% w/w), nicotinamide riboside (NR; 0.3% w/w) or both (COMBI) for 21 weeks. L-carnitine plasma levels were reduced by HFD and normalized by LC. NR supplementation raised its plasma metabolite levels demonstrating effective delivery. Although food intake and ambulatory activity were comparable in all groups, COMBI treatment significantly attenuated HFD-induced body weight gain, fat mass gain (-17%) and hepatic steatosis (-22%). Also, NR and COMBI reduced hepatic 4-hydroxynonenal adducts. Upstream-regulator gene analysis demonstrated that COMBI reversed detrimental effects of HFD on liver metabolism pathways and associated regulators, e.g., ACOX, SCAP, SREBF, PPARGC1B, and INSR. Combination treatment with LC and NR exerts protective effects on metabolic pathways and constitutes a new approach to attenuate HFD-induced obesity and NAFLD.


Asunto(s)
Carnitina/farmacología , Hígado Graso/metabolismo , Niacinamida/análogos & derivados , Obesidad/metabolismo , Animales , Biomarcadores , Modelos Animales de Enfermedad , Metabolismo Energético/efectos de los fármacos , Hígado Graso/tratamiento farmacológico , Hígado Graso/genética , Regulación de la Expresión Génica , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Niacinamida/farmacología , Obesidad/tratamiento farmacológico , Obesidad/genética , Estrés Oxidativo , Compuestos de Piridinio , Transducción de Señal
3.
Metabolism ; 97: 57-67, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31153978

RESUMEN

BACKGROUND: Muscle atrophy is defined as decreased muscle mass, associated with aging as well as with various chronic diseases and is a fundamental cause of frailty, functional decline and disability. Frailty represents a huge potential public health issue worldwide with high impact on healthcare costs. A major clinical issue is therefore to devise new strategies preventing muscle atrophy. In this study, we tested the efficacy of Vital01, a novel oral nutritional supplement (ONS), on body weight and muscle mass using a caloric restriction-induced mouse model for muscle atrophy. METHODS: Mice were calorically restricted for 2 weeks to induce muscle atrophy: one control group received 60% kcal of the normal chow diet and one intervention group received 30% kcal chow and 30 kcal% Vital01. The effects on body weight, lean body mass, muscle histology and transcriptome were assessed. In addition, the effects of Vital01, in mice with established muscle atrophy, were assessed and compared to a standard ONS. To this end, mice were first calorically restricted on a 60% kcal chow diet and then refed with either 100 kcal% chow, a mix of Vital01 (receiving 60% kcal chow and 40 kcal% Vital01) or with a mix of standard, widely prescribed ONS (receiving 60 kcal% chow and 40 kcal% Fortisip Compact). RESULTS: Vital01 attenuated weight loss (-15% weight loss for Vital01 vs. -25% for control group, p < 0.01) and loss of muscle mass (Vital01 with -13%, -12% and -18%, respectively, for gastrocnemius, quadriceps and tibialis vs. 25%, -23% and -28%, respectively, for control group, all p < 0.05) and also restored body weight, fat and muscle mass more efficiently when compared to Fortisip Compact. As assessed by transcriptome analysis and Western blotting of key proteins (e.g. phospoAKT, mTOR and S6K), Vital01 attenuated the catabolic and anabolic signaling pathways induced by caloric restriction and modulated inflammatory and mitochondrial pathways. In addition, Vital01 affected pathways related to matrix proteins/collagens homeostasis and tended to reduce caloric restriction-induced collagen fiber density in the quadriceps (with -27%, p = 0.051). CONCLUSIONS: We demonstrate that Vital01 preserves muscle mass in a calorically restricted mouse model for muscle atrophy. Vital01 had preventive effects when administered during development of muscle atrophy. Furthermore, when administered in a therapeutic setting to mice with established muscle atrophy, Vital01 rapidly restored body weight and accelerated the recurrence of fat and lean body mass more efficiently than Fortisip Compact. Bioinformatics analysis of gene expression data identified regulatory pathways that were specifically influenced by Vital01 in muscle.


Asunto(s)
Peso Corporal/fisiología , Músculo Esquelético/fisiología , Atrofia Muscular/fisiopatología , Animales , Composición Corporal/fisiología , Índice de Masa Corporal , Restricción Calórica/métodos , Suplementos Dietéticos , Modelos Animales de Enfermedad , Ingestión de Energía/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Pérdida de Peso/fisiología
4.
Front Immunol ; 10: 231, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30828334

RESUMEN

Despite scientific advances it remains difficult to predict the risk and benefit balance of immune interventions. Since a few years, network models have been built based on comprehensive datasets at multiple molecular/cellular levels (genes, gene products, metabolic intermediates, macromolecules, cells) to illuminate functional and structural relationships. Here we used a systems biology approach to identify key immune pathways involved in immune health endpoints and rank crucial candidate biomarkers to predict adverse and beneficial effects of nutritional immune interventions. First, a literature search was performed to select the molecular and cellular dynamics involved in hypersensitivity, autoimmunity and resistance to infection and cancer. Thereafter, molecular interaction between molecules and immune health endpoints was defined by connecting their relations by using database information. MeSH terms related to the immune health endpoints were selected resulting in the following selection: hypersensitivity (D006967: 184 genes), autoimmunity (D001327: 564 genes), infection (parasitic, bacterial, fungal and viral: 357 genes), and cancer (D009369: 3173 genes). In addition, a sequence of key processes was determined using Gene Ontology which drives the development of immune health disturbances resulting in the following selection: hypersensitivity (164 processes), autoimmunity (203 processes), infection (187 processes), and cancer (309 processes). Finally, an evaluation of the genes for each of the immune health endpoints was performed, which indicated that many genes played a role in multiple immune health endpoints, but also unique genes were observed for each immune health endpoint. This approach helps to build a screening/prediction tool which indicates the interaction of chemicals or food substances with immune health endpoint-related genes and suggests candidate biomarkers to evaluate risks and benefits. Several anti-cancer drugs and omega 3 fatty acids were evaluated as in silico test cases. To conclude, here we provide a systems biology approach to identify genes/molecules and their interaction with immune related disorders. Our examples illustrate that the prediction with our systems biology approach is promising and can be used to find both negatively and positively correlated interactions. This enables identification of candidate biomarkers to monitor safety and efficacy of therapeutic immune interventions.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/diagnóstico , Inmunoterapia/métodos , Biología de Sistemas/métodos , Animales , Biomarcadores/metabolismo , Humanos , Pronóstico , Resultado del Tratamiento
5.
Atherosclerosis ; 250: 158-65, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27236706

RESUMEN

BACKGROUND AND AIMS: Besides LDL-cholesterol, local vascular inflammation plays a key role in atherogenesis. Efficient therapies to treat the inflammatory component of the disease have not been established. The discovery of specialized inflammation-resolving mediators, such as resolvins may provide new opportunities for treatment. This study examines whether the ω-3 fatty acid eicosapentaenoic acid-derived resolvin E1 (RvE1), can reduce atherosclerosis, when administered alone or in combination with a cholesterol-lowering statin. METHODS: ApoE*3Leiden mice were fed a hypercholesterolemic diet for 9 weeks and subsequently treated with RvE1-low (1 mg/kg/day), RvE1-high (5 mg/kg/day), atorvastatin (1.5 mg/kg/day) or the combination of atorvastatin and RvE1-low for the following 16 weeks. RESULTS: RvE1-low and RvE1-high reduced atherosclerotic lesion size to the same extent (-35%; p < 0.05), attenuated the formation of severe lesions, also seen as a proportional increase in the presence of mild lesions, but did not alter plasma cholesterol levels. Cholesterol-lowering atorvastatin reduced atherosclerosis (-27%, p < 0.05), and the combination of RvE1 and atorvastatin further attenuated lesion size (-51%, p < 0.01) and increased the content of mild lesions. RvE1 did not affect plasma SAA, E-selectin, VCAM-1 or MCP-1 but did reduce plasma EPHX4 and down-regulated the local expression of pro-atherogenic genes in the aortae, (e.g. Cd74, Cd44, Ccl2, Ccr5 and Adam17) and significantly inactivated IFN-γ (p < 0.001) and TNF-α (p < 0.001) signalling pathways. CONCLUSIONS: RvE1 attenuates atherogenesis both alone and on top of a statin. The local effects of RvE1 are demonstrated by the modulated aortic expression of genes involved in inflammatory and immune responses, without altering plasma cholesterol or circulating SAA.


Asunto(s)
Aorta/patología , Atorvastatina/farmacología , Colesterol/sangre , Ácido Eicosapentaenoico/análogos & derivados , Lípidos/sangre , Animales , Aterosclerosis/sangre , LDL-Colesterol/sangre , Selectina E/sangre , Ácido Eicosapentaenoico/metabolismo , Ácido Eicosapentaenoico/farmacología , Ácidos Grasos Omega-3/metabolismo , Femenino , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Inflamación , Ratones , Ratones Noqueados para ApoE , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteína Amiloide A Sérica/metabolismo , Resultado del Tratamiento , Molécula 1 de Adhesión Celular Vascular/sangre
6.
J Lipid Res ; 56(11): 2085-93, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26342106

RESUMEN

Recently, we showed in APOE*3-Leiden cholesteryl ester transfer protein (E3L.CETP) mice that anacetrapib attenuated atherosclerosis development by reducing (V)LDL cholesterol [(V)LDL-C] rather than by raising HDL cholesterol. Here, we investigated the mechanism by which anacetrapib reduces (V)LDL-C and whether this effect was dependent on the inhibition of CETP. E3L.CETP mice were fed a Western-type diet alone or supplemented with anacetrapib (30 mg/kg body weight per day). Microarray analyses of livers revealed downregulation of the cholesterol biosynthesis pathway (P < 0.001) and predicted downregulation of pathways controlled by sterol regulatory element-binding proteins 1 and 2 (z-scores -2.56 and -2.90, respectively; both P < 0.001). These data suggest increased supply of cholesterol to the liver. We found that hepatic proprotein convertase subtilisin/kexin type 9 (Pcsk9) expression was decreased (-28%, P < 0.01), accompanied by decreased plasma PCSK9 levels (-47%, P < 0.001) and increased hepatic LDL receptor (LDLr) content (+64%, P < 0.01). Consistent with this, anacetrapib increased the clearance and hepatic uptake (+25%, P < 0.001) of [(14)C]cholesteryl oleate-labeled VLDL-mimicking particles. In E3L mice that do not express CETP, anacetrapib still decreased (V)LDL-C and plasma PCSK9 levels, indicating that these effects were independent of CETP inhibition. We conclude that anacetrapib reduces (V)LDL-C by two mechanisms: 1) inhibition of CETP activity, resulting in remodeled VLDL particles that are more susceptible to hepatic uptake; and 2) a CETP-independent reduction of plasma PCSK9 levels that has the potential to increase LDLr-mediated hepatic remnant clearance.


Asunto(s)
VLDL-Colesterol/sangre , Dislipidemias/sangre , Hipolipemiantes/farmacología , Oxazolidinonas/farmacología , Proproteína Convertasas/sangre , Serina Endopeptidasas/sangre , Animales , Enfermedades Cardiovasculares/prevención & control , Proteínas de Transferencia de Ésteres de Colesterol/antagonistas & inhibidores , Proteínas de Transferencia de Ésteres de Colesterol/metabolismo , Regulación hacia Abajo , Evaluación Preclínica de Medicamentos , Dislipidemias/tratamiento farmacológico , Dislipidemias/enzimología , Femenino , Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes , Hipolipemiantes/uso terapéutico , Metabolismo de los Lípidos/efectos de los fármacos , Redes y Vías Metabólicas , Ratones Transgénicos , Oxazolidinonas/uso terapéutico , Proproteína Convertasa 9
7.
J Hepatol ; 62(5): 1180-6, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25514555

RESUMEN

BACKGROUND & AIMS: Anthocyanins may have beneficial effects on lipid metabolism and inflammation and are demonstrated to have hepatoprotective properties in models of restraint-stress- and chemically-induced liver damage. However, their potential to protect against non-alcoholic steatohepatitis (NASH) under conditions relevant for human pathogenesis remains unclear. Therefore, we studied the effects of the standardised anthocyanin-rich extract Mirtoselect on diet-induced NASH in a translational model of disease. METHODS: ApoE(∗)3Leiden mice were fed a Western-type cholesterol-containing diet without (HC) or with 0.1% (w/w) Mirtoselect (HCM) for 20weeks to study the effects on diet-induced NASH. RESULTS: Mirtoselect attenuated HC-induced hepatic steatosis, as observed by decreased macro- and microvesicular hepatocellular lipid accumulation and reduced hepatic cholesteryl ester content. This anti-steatotic effect was accompanied by local anti-inflammatory effects in liver, as demonstrated by reduced inflammatory cell clusters and reduced neutrophil infiltration in HCM. On a molecular level, HC diet significantly induced hepatic expression of pro-inflammatory genes Tnf, Emr1, Ccl2, Mpo, Cxcl1, and Cxcl2 while this induction was less pronounced or significantly decreased in HCM. A similar quenching effect was observed for HC-induced pro-fibrotic genes, Acta2 and Col1a1 and this anti-fibrotic effect of Mirtoselect was confirmed histologically. Many of the pro-inflammatory and pro-fibrotic parameters positively correlated with intrahepatic free cholesterol levels. Mirtoselect significantly reduced accumulation and crystallisation of intrahepatic free cholesterol, providing a possible mechanism for the observed hepatoprotective effects. CONCLUSIONS: Mirtoselect attenuates development of NASH, reducing hepatic lipid accumulation, inflammation and fibrosis, possibly mediated by local anti-inflammatory effects associated with reduced accumulation and crystallisation of intrahepatic free cholesterol.


Asunto(s)
Antocianinas/farmacología , Cirrosis Hepática/prevención & control , Enfermedad del Hígado Graso no Alcohólico , Vaccinium myrtillus/química , Actinas/metabolismo , Animales , Antiinfecciosos/farmacología , Proteínas de Unión al Calcio , Quimiocina CXCL1/metabolismo , Ésteres del Colesterol/metabolismo , Colesterol en la Dieta/metabolismo , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Citoprotección , Dieta Occidental , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/etiología , Cirrosis Hepática/fisiopatología , Ratones , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/fisiopatología , Extractos Vegetales , Receptores de Superficie Celular/metabolismo , Receptores Acoplados a Proteínas G , Resultado del Tratamiento
8.
Pharmacogenet Genomics ; 22(12): 837-45, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23086299

RESUMEN

AIMS: Combination-drug therapy takes advantage of the complementary action of their individual components, thereby potentiating its therapeutic effect. Potential disadvantages include side effects that are not foreseen on basis of the data available from drug monotherapy. Here, we used a systems biology approach to understand both the efficacy and the side effects of a cholesterol-lowering drug-combination therapy on the basis of the biological pathways and molecular processes affected by each drug alone or in combination. METHODS AND RESULTS: ApoE*3Leiden transgenic mice, a mouse model with human-like cholesterol-lowering drug responses, were treated with rosuvastatin and ezetimibe, alone and in combination. Analyses included functional responses, viz. effects on cardiovascular risk factors, inflammation, and atherosclerosis, and measurement of global gene expression, and identification of enriched biological pathways and molecular processes. Combination therapy reduced plasma cholesterol, plasma inflammation markers, and atherosclerosis stronger than the single drugs did. Systems biology analysis at the level of biological processes shows that the therapeutic benefit of combined therapy is largely the result of additivity of the complementary mechanisms of action of the two single drugs. Importantly, combination therapy also exerted a significant effect on 16 additional and mostly NF-κB-linked signaling processes, 11 of which tended to be regulated in a similar direction with monotherapy. CONCLUSION: This study shows that gene expression analysis together with bioinformatics pathway analysis has the potential to help predict and identify drug combination-specific complementary and side effects.


Asunto(s)
Anticolesterolemiantes/uso terapéutico , Azetidinas/uso terapéutico , Fluorobencenos/uso terapéutico , Pirimidinas/uso terapéutico , Sulfonamidas/uso terapéutico , Animales , Anticolesterolemiantes/administración & dosificación , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Aterosclerosis/tratamiento farmacológico , Azetidinas/administración & dosificación , Quimioterapia Combinada , Ezetimiba , Femenino , Fluorobencenos/administración & dosificación , Ratones , Ratones Transgénicos , Pirimidinas/administración & dosificación , Factores de Riesgo , Rosuvastatina Cálcica , Sulfonamidas/administración & dosificación , Biología de Sistemas
9.
Mol Nutr Food Res ; 56(7): 1081-9, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22611002

RESUMEN

SCOPE: This study addresses whether early life arachidonic acid (ARA)/docosahexaenoic acid (DHA) supplementation or eicosapentaenoic acid (EPA)/DHA (Omacor) supplementation affects body weight gain, lipid metabolism, and adipose tissue quantity and quality in later life in ApoE*3Leiden-transgenic mice, a humanized model for hyperlipidemia and mild obesity. METHODS AND RESULTS: Four-week-old male ApoE*3Leiden mice were fed chow diet with or without a mixture of ARA (0.129 wt%) and DHA (0.088 wt%) or Omacor (0.30 wt% EPA, 0.25 wt% DHA). At age 12 weeks, mice were fed high-fat/high-carbohydrate (HFHC) diet without above supplements until age 20 weeks. Control mice received chow diet throughout the study. Mice receiving ARA/DHA gained less body weight compared to control and this effect was sustained when fed HFHC. Omacor had no significant effect on body weight gain. Plasma cholesterol and triglycerides were significantly lowered by both supplementations. At 20 weeks, epididymal fat mass was less in ARA/DHA-supplemented mice, while Omacor had no significant effect on fat mass. Both ARA/DHA and Omacor reduced inguinal adipocyte cell size; only ARA/DHA significantly reduced epididymal macrophage infiltration. CONCLUSION: This study shows that early life ARA/DHA, but not Omacor supplementation improves body weight gain later in life. ARA/DHA and to a lesser extent Omacor both improved adipose tissue quality.


Asunto(s)
Adiposidad , Fármacos Antiobesidad/uso terapéutico , Ácido Araquidónico/uso terapéutico , Ácidos Docosahexaenoicos/uso terapéutico , Hiperlipidemias/prevención & control , Hipolipemiantes/uso terapéutico , Obesidad/prevención & control , Tejido Adiposo Blanco/inmunología , Tejido Adiposo Blanco/patología , Animales , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Tamaño de la Célula , Colesterol/sangre , Suplementos Dietéticos , Hiperlipidemias/sangre , Hiperlipidemias/inmunología , Hiperlipidemias/patología , Macrófagos/inmunología , Macrófagos/patología , Masculino , Ratones , Ratones Transgénicos , Obesidad/sangre , Obesidad/inmunología , Obesidad/patología , Organismos Libres de Patógenos Específicos , Triglicéridos/sangre , Aumento de Peso
10.
J Nutr ; 141(5): 863-9, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21411607

RESUMEN

Chronic inflammation and proatherogenic lipids are important risk factors of cardiovascular disease (CVD). Specific dietary constituents such as polyphenols and fish oils may improve cardiovascular risk factors and may have a beneficial effect on disease outcomes. We hypothesized that the intake of an antiinflammatory dietary mixture (AIDM) containing resveratrol, lycopene, catechin, vitamins E and C, and fish oil would reduce inflammatory risk factors, proatherogenic lipids, and endpoint atherosclerosis. AIDM was evaluated in an inflammation model, male human C-reactive protein (CRP) transgenic mice, and an atherosclerosis model, female ApoE*3Leiden transgenic mice. Two groups of male human-CRP transgenic mice were fed AIDM [0.567% (wt:wt) powder and 0.933% (wt:wt oil)] or placebo for 6 wk. The effects of AIDM on basal and IL-1ß-stimulated CRP expression were investigated. AIDM reduced cytokine-induced human CRP and fibrinogen expression in human-CRP transgenic mice. In the atherosclerosis study, 2 groups of female ApoE*3Leiden transgenic mice were fed an atherogenic diet supplemented with AIDM [0.567% (wt:wt) powder and 0.933% (wt:wt oil)] or placebo for 16 wk. AIDM strongly reduced plasma cholesterol, TG, and serum amyloid A concentrations compared with placebo. Importantly, long-term treatment of ApoE*3Leiden mice with AIDM markedly reduced the development of atherosclerosis by 96% compared with placebo. The effect on atherosclerosis was paralleled by a reduced expression of the vascular inflammation markers and adhesion molecules inter-cellular adhesion molecule-1 and E-selectin. Dietary supplementation of AIDM improves lipid and inflammatory risk factors of CVD and strongly reduces atherosclerotic lesion development in female transgenic mice.


Asunto(s)
Antioxidantes/uso terapéutico , Aterosclerosis/dietoterapia , Dieta , Aceites de Pescado/uso terapéutico , Proteínas de Fase Aguda/análisis , Animales , Antioxidantes/administración & dosificación , Apolipoproteína E3/genética , Ácido Ascórbico/administración & dosificación , Ácido Ascórbico/uso terapéutico , Aterosclerosis/sangre , Aterosclerosis/epidemiología , Aterosclerosis/patología , Biomarcadores/sangre , Proteína C-Reactiva/análisis , Proteína C-Reactiva/genética , Carotenoides/administración & dosificación , Carotenoides/uso terapéutico , Catequina/administración & dosificación , Catequina/uso terapéutico , Femenino , Aceites de Pescado/administración & dosificación , Humanos , Licopeno , Masculino , Ratones , Ratones Transgénicos , Resveratrol , Factores de Riesgo , Estilbenos/administración & dosificación , Estilbenos/uso terapéutico , Vitamina E/administración & dosificación , Vitamina E/uso terapéutico
11.
Arterioscler Thromb Vasc Biol ; 25(1): 161-7, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15514207

RESUMEN

OBJECTIVE: To evaluate whether low-dose atorvastatin suppresses atherosclerotic lesion progression and inflammation in apolipoprotein E*3 (apoE*3)-Leiden mice beyond its cholesterol-lowering effect. METHODS AND RESULTS: ApoE*3-Leiden mice were fed a high-cholesterol (HC) diet until mild atherosclerotic lesions had formed. Subsequently, HC diet feeding was continued or mice received HC supplemented with 0.002% (w/w) atorvastatin (HC+A), resulting in 19% plasma cholesterol lowering, or mice received a low-cholesterol (LC) diet to establish a plasma cholesterol level similar to that achieved in the HC+A group. HC+A and LC diet reduced, significantly and to the same extent, lesion progression and complication in the aortic root, as assessed by measuring total atherosclerotic lesion area, lesion severity, and macrophage and smooth muscle cell area. In the aortic arch, HC+A but not LC blocked lesion progression. HC+A and LC reduced vascular inflammation (ie, expression of macrophage migration inhibitory factor , plasminogen activator inhibitor- 1, matrix metalloproteinase-9), but HC+A additionally suppressed vascular cell adhesion molecule-1 expression and, in parallel, monocyte adhesion. In contrast, low-dose atorvastatin showed no antiinflammatory action toward hepatic inflammation markers (serum amyloid A, C-reactive protein [CRP]) in apoE*3-Leiden mice and human CRP transgenic mice. CONCLUSIONS: Low-dose atorvastatin cholesterol-dependently reduces lesion progression in the aortic root but shows antiinflammatory vascular activity and tends to retard atherogenesis in the aortic arch beyond its cholesterol-lowering effect.


Asunto(s)
Apolipoproteínas E/genética , Arteriosclerosis/dietoterapia , Arteriosclerosis/tratamiento farmacológico , Ácidos Heptanoicos/farmacología , Pirroles/farmacología , Animales , Anticolesterolemiantes/administración & dosificación , Anticolesterolemiantes/farmacología , Anticolesterolemiantes/uso terapéutico , Aorta/patología , Apolipoproteína E3 , Arteriosclerosis/patología , Atorvastatina , Biomarcadores/metabolismo , Proteína C-Reactiva/metabolismo , Colesterol/sangre , Dieta , Dieta Aterogénica , Esquema de Medicación , Femenino , Ácidos Heptanoicos/administración & dosificación , Ácidos Heptanoicos/uso terapéutico , Inflamación/dietoterapia , Inflamación/tratamiento farmacológico , Masculino , Ratones , Ratones Transgénicos , Pirroles/administración & dosificación , Pirroles/uso terapéutico , Proteína Amiloide A Sérica/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA