Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
Kidney Int ; 90(1): 181-91, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27188504

RESUMO

Delayed graft function (DGF) following kidney transplantation affects long-term graft function and survival and is considered a manifestation of ischemia reperfusion injury. Preclinical studies characterize metabolic defects resulting from mitochondrial damage as primary driver of ischemia reperfusion injury. In a comprehensive approach that included sequential establishment of postreperfusion arteriovenous concentration differences over the human graft, metabolomic and genomic analysis in tissue biopsies taken before and after reperfusion, we tested whether the preclinical observations translate to the context of clinical DGF. This report is based on sequential studies of 66 eligible patients of which 22 experienced DGF. Grafts with no DGF immediately recovered aerobic respiration as indicated by prompt cessation of lactate release following reperfusion. In contrast, grafts with DGF failed to recover aerobic respiration and showed persistent adenosine triphosphate catabolism indicated by a significant persistently low post reperfusion tissue glucose-lactate ratio and continued significant post-reperfusion lactate and hypoxanthine release (net arteriovenous difference for lactate and hypoxanthine at 30 minutes). The metabolic data for the group with DGF point to a persistent post reperfusion mitochondrial defect, confirmed by functional (respirometry) and morphological analyses. The archetypical mitochondrial stabilizing peptide SS-31 significantly preserved mitochondrial function in human kidney biopsies following simulated ischemia reperfusion. Thus, development of DGF is preceded by a profound post-reperfusion metabolic deficit resulting from severe mitochondrial damage. Strategies aimed at preventing DGF should be focused on safeguarding a minimally required post-reperfusion metabolic competence.


Assuntos
Aloenxertos/patologia , Função Retardada do Enxerto/metabolismo , Sobrevivência de Enxerto , Transplante de Rim/efeitos adversos , Mitocôndrias/patologia , Traumatismo por Reperfusão/complicações , Aloenxertos/metabolismo , Biópsia , Estudos de Coortes , Função Retardada do Enxerto/epidemiologia , Função Retardada do Enxerto/etiologia , Função Retardada do Enxerto/patologia , Feminino , Humanos , Incidência , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Masculino , Pessoa de Meia-Idade , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Oligopeptídeos/uso terapêutico , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia
2.
FASEB J ; 29(11): 4600-13, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26198450

RESUMO

Metabolism maintains homeostasis at chronic hypercaloric conditions, activating postprandial response mechanisms, which come at the cost of adaptation processes such as energy storage, eventually with negative health consequences. This study quantified the metabolic adaptation capacity by studying challenge response curves. After a high-fat challenge, the 8 h response curves of 61 biomarkers related to adipose tissue mass and function, systemic stress, metabolic flexibility, vascular health, and glucose metabolism was compared between 3 metabolic health stages: 10 healthy men, before and after 4 wk of high-fat, high-calorie diet (1300 kcal/d extra), and 9 men with metabolic syndrome (MetS). The MetS subjects had increased fasting concentrations of biomarkers representing the 3 core processes, glucose, TG, and inflammation control, and the challenge response curves of most biomarkers were altered. After the 4 wk hypercaloric dietary intervention, these 3 processes were not changed, as compared with the preintervention state in the healthy subjects, whereas the challenge response curves of almost all endocrine, metabolic, and inflammatory processes regulating these core processes were altered, demonstrating major molecular physiologic efforts to maintain homeostasis. This study thus demonstrates that change in challenge response is a more sensitive biomarker of metabolic resilience than are changes in fasting concentrations.


Assuntos
Tecido Adiposo/metabolismo , Glicemia/metabolismo , Gorduras na Dieta/administração & dosagem , Homeostase/efeitos dos fármacos , Triglicerídeos/sangue , Adulto , Idoso , Biomarcadores/sangue , Humanos , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Fatores de Tempo
3.
J Hepatol ; 62(5): 1180-6, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25514555

RESUMO

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.


Assuntos
Antocianinas/farmacologia , Cirrose Hepática/prevenção & controle , Hepatopatia Gordurosa não Alcoólica , Vaccinium myrtillus/química , Actinas/metabolismo , Animais , Anti-Infecciosos/farmacologia , Proteínas de Ligação ao Cálcio , Quimiocina CXCL1/metabolismo , Ésteres do Colesterol/metabolismo , Colesterol na Dieta/metabolismo , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Citoproteção , Dieta Ocidental , Humanos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Fígado/metabolismo , Fígado/patologia , Cirrose Hepática/etiologia , Cirrose Hepática/fisiopatologia , Camundongos , Hepatopatia Gordurosa não Alcoólica/complicações , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Hepatopatia Gordurosa não Alcoólica/fisiopatologia , Extratos Vegetais , Receptores de Superfície Celular/metabolismo , Receptores Acoplados a Proteínas G , Resultado do Tratamento
4.
Nat Med ; 13(5): 587-96, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17435771

RESUMO

The cytokine macrophage migration inhibitory factor (MIF) plays a critical role in inflammatory diseases and atherogenesis. We identify the chemokine receptors CXCR2 and CXCR4 as functional receptors for MIF. MIF triggered G(alphai)- and integrin-dependent arrest and chemotaxis of monocytes and T cells, rapid integrin activation and calcium influx through CXCR2 or CXCR4. MIF competed with cognate ligands for CXCR4 and CXCR2 binding, and directly bound to CXCR2. CXCR2 and CD74 formed a receptor complex, and monocyte arrest elicited by MIF in inflamed or atherosclerotic arteries involved both CXCR2 and CD74. In vivo, Mif deficiency impaired monocyte adhesion to the arterial wall in atherosclerosis-prone mice, and MIF-induced leukocyte recruitment required Il8rb (which encodes Cxcr2). Blockade of Mif but not of canonical ligands of Cxcr2 or Cxcr4 in mice with advanced atherosclerosis led to plaque regression and reduced monocyte and T-cell content in plaques. By activating both CXCR2 and CXCR4, MIF displays chemokine-like functions and acts as a major regulator of inflammatory cell recruitment and atherogenesis. Targeting MIF in individuals with manifest atherosclerosis can potentially be used to treat this condition.


Assuntos
Aterosclerose/fisiopatologia , Endotélio Vascular/fisiologia , Inflamação/fisiopatologia , Fatores Inibidores da Migração de Macrófagos/fisiologia , Receptores CXCR4/fisiologia , Receptores de Interleucina-8B/fisiologia , Aorta , Quimiotaxia , Humanos , Leucócitos/fisiologia , Ligantes , Fatores Inibidores da Migração de Macrófagos/farmacologia , Monócitos/fisiologia , Linfócitos T/fisiologia
5.
Br J Nutr ; 110(1): 77-85, 2013 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-23211714

RESUMO

A high-fat diet disturbs the composition and function of the gut microbiota and generates local gut-associated and also systemic responses. Intestinal mast cells, for their part, secrete mediators which play a role in the orchestration of physiological and immunological functions of the intestine. Probiotic bacteria, again, help to maintain the homeostasis of the gut microbiota by protecting the gut epithelium and regulating the local immune system. In the present study, we explored the effects of two probiotic bacteria, Lactobacillus rhamnosus GG (GG) and Propionibacterium freudenreichii spp. shermanii JS (PJS), on high fat-fed ApoE*3Leiden mice by estimating the mast cell numbers and the immunoreactivity of TNF-α and IL-10 in the intestine, as well as plasma levels of several markers of inflammation and parameters of lipid metabolism. We found that mice that received GG and PJS exhibited significantly lower numbers of intestinal mast cells compared with control mice. PJS lowered intestinal immunoreactivity of TNF-α, while GG increased intestinal IL-10. PJS was also observed to lower the plasma levels of markers of inflammation including vascular cell adhesion molecule 1, and also the amount of gonadal adipose tissue. GG lowered alanine aminotransferase, a marker of hepatocellular activation. Collectively, these data demonstrate that probiotic GG and PJS tend to down-regulate both intestinal and systemic pro-inflammatory changes induced by a high-fat diet in this humanised mouse model.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Inflamação/prevenção & controle , Mucosa Intestinal/microbiologia , Lacticaseibacillus rhamnosus , Mastócitos/metabolismo , Probióticos/uso terapêutico , Propionibacterium , Tecido Adiposo/metabolismo , Alanina Transaminase/sangue , Animais , Gônadas/metabolismo , Inflamação/etiologia , Inflamação/imunologia , Inflamação/metabolismo , Mediadores da Inflamação/sangue , Interleucina-10/metabolismo , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Metabolismo dos Lipídeos , Fígado/efeitos dos fármacos , Fígado/enzimologia , Masculino , Metagenoma , Camundongos , Camundongos Endogâmicos , Fator de Necrose Tumoral alfa/metabolismo , Molécula 1 de Adesão de Célula Vascular/sangue
6.
Physiol Genomics ; 44(5): 293-304, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22234995

RESUMO

The prevalence of diabetes mellitus Type 2 could be significantly reduced by early identification of subjects at risk, allowing for better prevention and earlier treatment. Glucose intolerance (GI) is a hallmark of the prediabetic stage. This study aims at identifying 1) prognostic biomarkers predicting the risk of developing GI later in life and 2) diagnostic biomarkers reflecting the degree of already manifest GI. To this end, disease development was followed over time in mice, and biomarkers were identified using lipidomics and transcriptomics. Young adult ApoE3Leiden mice were treated a high-fat diet for 12 wk to induce GI. Blood was collected before and during disease development. The individual extent of GI was determined with a glucose tolerance test and the area under the curve (AUC) was calculated for each animal. Subject-specific AUC values were correlated to the plasma lipidome (t = 0) and the white blood cell (WBC) transcriptome (t = 0, 6, and 12 wk) to identify prognostic and diagnostic biomarkers, respectively. The plasma ratio of specific free fatty acids prior to high-fat feeding (C16:1/C16:0, C18:1/C18:0 and C18:2/C22:6) was significantly correlated with the AUC and predictive for future GI. Subsequently, the expression level of specific WBC genes (Acss2, Arfgap1, Tfrc, Cox6b2, Barhl2, Abcb4, Cyp4b1, Sars2, Fgf16, and Tceal8) reflected the individual degree of GI during disease progression. Specific plasma free fatty acids as well as their ratio can be used to predict future GI. The expression levels of specific WBC genes can serve as easy accessible markers to diagnose and monitor already existing GI.


Assuntos
Apolipoproteína E3/genética , Biomarcadores/análise , Intolerância à Glucose/diagnóstico , Animais , Biomarcadores/sangue , Biomarcadores/metabolismo , Perfilação da Expressão Gênica , Intolerância à Glucose/sangue , Intolerância à Glucose/genética , Leucócitos/química , Leucócitos/metabolismo , Lipídeos/análise , Lipídeos/sangue , Masculino , Metaboloma , Camundongos , Camundongos Transgênicos , Análise em Microsséries , Técnicas de Diagnóstico Molecular , Prognóstico , Transcriptoma , Estudos de Validação como Assunto
7.
Pharmacogenet Genomics ; 22(12): 837-45, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23086299

RESUMO

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.


Assuntos
Anticolesterolemiantes/uso terapêutico , Azetidinas/uso terapêutico , Fluorbenzenos/uso terapêutico , Pirimidinas/uso terapêutico , Sulfonamidas/uso terapêutico , Animais , Anticolesterolemiantes/administração & dosagem , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Aterosclerose/tratamento farmacológico , Azetidinas/administração & dosagem , Quimioterapia Combinada , Ezetimiba , Feminino , Fluorbenzenos/administração & dosagem , Camundongos , Camundongos Transgênicos , Pirimidinas/administração & dosagem , Fatores de Risco , Rosuvastatina Cálcica , Sulfonamidas/administração & dosagem , Biologia de Sistemas
8.
Mol Med ; 17(3-4): 189-93, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21103669

RESUMO

The possible contribution of tumor necrosis factor-α (TNF-α) to the development of obesity-associated insulin resistance in humans is still controversial. Our study investigated the effect of TNF-α neutralization on insulin resistance in healthy, obese and insulin resistant men. We performed a prospective, randomized, double-blind placebo-controlled trial in nine young, healthy obese male subjects with metabolic syndrome and insulin resistance. Volunteers received three infusions (wks 0, 2 and 6) of infliximab or placebo. Insulin resistance was measured at baseline and after 70 d by homeostatic model assessment (HOMA) index as well as by minimal model analysis of an intravenous glucose tolerance test. Endothelial function was accessed before and after intervention by flow mediated dilation. Infliximab improved the inflammatory status as indicated by reduced high sensitivity C-reactive protein (hsCRP) and fibrinogen levels (2.77 ± 0.6 to 1.8 ± 0.5 µg/L, and 3.42 ± 0.18 to 3.18 ± 0.28 g/L; (day 0 and day 70, P = 0.020 and 0.037 respectively), but did not improve insulin resistance (HOMA index and intravenous glucose-tolerance test [ivGGT]) or endothelial function. Despite improvements in inflammatory status, chronic TNF-α neutralization does not improve insulin resistance or endothelial function in seemingly healthy, but obese, insulin-resistant volunteers. This study severely questions the proposal that TNF-α is a causative link between adiposity and insulin resistance.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Endotélio Vascular/efeitos dos fármacos , Resistência à Insulina , Síndrome Metabólica/tratamento farmacológico , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Adulto , Anti-Inflamatórios/administração & dosagem , Anti-Inflamatórios/imunologia , Anti-Inflamatórios/uso terapêutico , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/imunologia , Proteína C-Reativa/metabolismo , Endotélio Vascular/fisiopatologia , Fibrinogênio/metabolismo , Teste de Tolerância a Glucose , Humanos , Infliximab , Infusões Intravenosas , Masculino , Síndrome Metabólica/sangue , Síndrome Metabólica/complicações , Obesidade/complicações , Estudos Prospectivos , Resultado do Tratamento , Fator de Necrose Tumoral alfa/imunologia
9.
J Nutr ; 141(5): 863-9, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21411607

RESUMO

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.


Assuntos
Antioxidantes/uso terapêutico , Aterosclerose/dietoterapia , Dieta , Óleos de Peixe/uso terapêutico , Proteínas de Fase Aguda/análise , Animais , Antioxidantes/administração & dosagem , Apolipoproteína E3/genética , Ácido Ascórbico/administração & dosagem , Ácido Ascórbico/uso terapêutico , Aterosclerose/sangue , Aterosclerose/epidemiologia , Aterosclerose/patologia , Biomarcadores/sangue , Proteína C-Reativa/análise , Proteína C-Reativa/genética , Carotenoides/administração & dosagem , Carotenoides/uso terapêutico , Catequina/administração & dosagem , Catequina/uso terapêutico , Feminino , Óleos de Peixe/administração & dosagem , Humanos , Licopeno , Masculino , Camundongos , Camundongos Transgênicos , Resveratrol , Fatores de Risco , Estilbenos/administração & dosagem , Estilbenos/uso terapêutico , Vitamina E/administração & dosagem , Vitamina E/uso terapêutico
10.
Circ Res ; 105(1): 99-107, 2009 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-19478200

RESUMO

Chronic inflammation in white adipose tissue (WAT) is positively associated with obesity, insulin resistance (IR) and the development of type 2 diabetes. The proinflammatory cytokine MIF (macrophage migration inhibitory factor) is an essential, upstream component of the inflammatory cascade. This study examines whether MIF is required for the development of obesity, IR, glucose intolerance, and atherosclerosis in the LDL receptor-deficient (Ldlr(-/-)) mouse model of disease. Ldlr(-/-) mice develop IR and glucose intolerance within 15 weeks, whereas Mif(-/-)Ldlr(-/-) littermates are protected. MIF deficiency does not affect obesity and lipid risk factors but specifically reduces inflammation in WAT and liver, as reflected by lower plasma serum amyloid A and fibrinogen levels at baseline and under inflammatory conditions. Conversely, MIF stimulates the in vivo expression of human C-reactive protein, an inflammation marker and risk factor of IR and cardiovascular disease. In WAT, MIF deficiency reduces nuclear c-Jun levels and improves insulin sensitivity; MIF deficiency also reduces macrophage accumulation in WAT and blunts the expression of two proteins that regulate macrophage infiltration (intercellular adhesion molecule-1, CD44). Mechanistic parallels to WAT were observed in aorta, where the absence of MIF reduces monocyte adhesion, macrophage lesion content, and atherosclerotic lesion size. These data highlight the physiological importance of chronic inflammation in development of IR and atherosclerosis and suggest that MIF is a potential therapeutic target for reducing the inflammatory component of metabolic and cardiovascular disorders.


Assuntos
Tecido Adiposo Branco/patologia , Inflamação/etiologia , Resistência à Insulina , Fatores Inibidores da Migração de Macrófagos/deficiência , Animais , Aterosclerose/etiologia , Biomarcadores/sangue , Doença Crônica , Intolerância à Glucose/etiologia , Humanos , Fígado/patologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Receptores de LDL/deficiência
11.
BMC Bioinformatics ; 10: 52, 2009 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-19200393

RESUMO

BACKGROUND: In the fields of life sciences, so-called designed studies are used for studying complex biological systems. The data derived from these studies comply with a study design aimed at generating relevant information while diminishing unwanted variation (noise). Knowledge about the study design can be used to decompose the total data into data blocks that are associated with specific effects. Subsequent statistical analysis can be improved by this decomposition if these are applied on selected combinations of effects. RESULTS: The benefit of this approach was demonstrated with an analysis that combines multivariate PLS (Partial Least Squares) regression with data decomposition from ANOVA (Analysis of Variance): ANOVA-PLS. As a case, a nutritional intervention study is used on Apoliprotein E3-Leiden (APOE3Leiden) transgenic mice to study the relation between liver lipidomics and a plasma inflammation marker, Serum Amyloid A. The ANOVA-PLS performance was compared to PLS regression on the non-decomposed data with respect to the quality of the modelled relation, model reliability, and interpretability. CONCLUSION: It was shown that ANOVA-PLS leads to a better statistical model that is more reliable and better interpretable compared to standard PLS analysis. From a following biological interpretation, more relevant metabolites were derived from the model. The concept of combining data composition with a subsequent statistical analysis, as in ANOVA-PLS, is however not limited to PLS regression in metabolomics but can be applied for many statistical methods and many different types of data.


Assuntos
Metabolômica/estatística & dados numéricos , Modelos Estatísticos , Animais , Bases de Dados Factuais , Humanos , Metabolômica/métodos
12.
Cardiovasc Res ; 79(3): 360-76, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18487233

RESUMO

In the past few years, inflammation has emerged as a major driving force of atherosclerotic lesion development. It is now well-established that from early lesion to vulnerable plaque formation, numerous cellular and molecular inflammatory components participate in the disease process. The most prominent cells that invade in evolving lesions are monocyte-derived macrophages and T-lymphocytes. Both cell types produce a wide array of soluble inflammatory mediators (cytokines, chemokines) which are critically important in the initiation and perpetuation of the disease. This review summarizes the currently available information from mouse studies on the contribution of a specified group of cytokines expressed in atherosclerotic lesions, viz. interleukins (IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-10, IL-12, IL-18, IL-20) and macrophage-associated cytokines [tumour necrosis factor-alpha (TNF-alpha); macrophage migration inhibitory factor (MIF); interferon-gamma (IFN-gamma); colony stimulating factors G-CSF,-M-CSF,-GM-CSF) to atherogenesis. Emphasis is put on the consistency of the effects of these cytokines, i.e. inasmuch an effect depends on the experimental approach applied (overexpression/deletion, strain, gender, dietary conditions, and disease stage). An important outcome of this survey is (i) that only for a few cytokines there is sufficient consistent data allowing classifying them as typically proatherogenic (IL-1, IL-12, IL-18, MIF, IFN-gamma, TNF-alpha, and M-CSF) or antiatherogenic (IL-10) and (ii) that some cytokines (IL-4, IL-6 and GM-CSF) can exert pro- or anti-atherogenic effects depending on the experimental conditions. This knowledge can be used for improved early detection, prevention and treatment of atherosclerosis.


Assuntos
Aterosclerose/imunologia , Citocinas/metabolismo , Inflamação/imunologia , Transdução de Sinais , Animais , Fatores Estimuladores de Colônias/metabolismo , Modelos Animais de Doenças , Interferon gama/metabolismo , Interleucinas/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , Camundongos , Fator de Necrose Tumoral alfa/metabolismo
13.
Arterioscler Thromb Vasc Biol ; 27(8): 1706-21, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17541027

RESUMO

Atherosclerosis is a multifactorial highly-complex disease with numerous etiologies that work synergistically to promote lesion development. The ability to develop preventive and ameliorative treatments will depend on animal models that mimic the human subject metabolically and pathophysiologically and will develop lesions comparable to those in humans. The mouse is the most useful, economic, and valid model for studying atherosclerosis and exploring effective therapeutic approaches. Among the most widely used mouse models for atherosclerosis are apolipoprotein E-deficient (ApoE-/-) and LDL receptor-deficient (LDLr-/-) mice. An up-and-coming model is the ApoE*3Leiden (E3L) transgenic mouse. Here, we review studies that have explored how and to what extent these mice respond to compounds directed at treatment of the risk factors hypercholesterolemia, hypertriglyceridemia, hypertension, and inflammation. An important outcome of this survey is that the different models used may differ markedly from one another in their response to a specific experimental manipulation. The choice of a model is therefore of critical importance and should take into account the risk factor to be studied and the working spectrum of the compounds tested.


Assuntos
Apolipoproteínas E/deficiência , Aterosclerose/tratamento farmacológico , Modelos Animais de Doenças , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Animais , Anti-Hipertensivos/farmacologia , Aterosclerose/fisiopatologia , Relação Dose-Resposta a Droga , Esquema de Medicação , Dislipidemias/tratamento farmacológico , Dislipidemias/fisiopatologia , Humanos , Hipertensão/tratamento farmacológico , Hipertensão/fisiopatologia , Hipolipemiantes/farmacologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Sensibilidade e Especificidade
14.
Artigo em Inglês | MEDLINE | ID: mdl-18511357

RESUMO

We report a sensitive, generic method for quantitative profiling of bile acids and other endogenous metabolites in small quantities of various biological fluids and tissues. The method is based on a straightforward sample preparation, separation by reversed-phase high performance liquid-chromatography mass spectrometry (HPLC-MS) and electrospray ionisation in the negative ionisation mode (ESI-). Detection is performed in full scan using the linear ion trap Fourier transform mass spectrometer (LTQ-FTMS) generating data for many (endogenous) metabolites, not only bile acids. A validation of the method in urine, plasma and liver was performed for 17 bile acids including their taurine, sulfate and glycine conjugates. The method is linear in the 0.01-1 microM range. The accuracy in human plasma ranges from 74 to 113%, in human urine 77 to 104% and in mouse liver 79 to 140%. The precision ranges from 2 to 20% for pooled samples even in studies with large number of samples (n>250). The method was successfully applied to a multi-compartmental APOE*3-Leiden mouse study, the main goal of which was to analyze the effect of increasing dietary cholesterol concentrations on hepatic cholesterol homeostasis and bile acid synthesis. Serum and liver samples from different treatment groups were profiled with the new method. Statistically significant differences between the diet groups were observed regarding total as well as individual bile acid concentrations.


Assuntos
Ácidos e Sais Biliares/análise , Cromatografia Líquida/métodos , Biologia Computacional/métodos , Espectrometria de Massas/métodos , Metabolismo , Animais , Ácidos e Sais Biliares/sangue , Ácidos e Sais Biliares/urina , Colesterol na Dieta/administração & dosagem , Colesterol na Dieta/farmacologia , Análise de Fourier , Humanos , Fígado/química , Fígado/efeitos dos fármacos , Camundongos , Reprodutibilidade dos Testes
15.
J Hypertens ; 25(12): 2454-62, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17984667

RESUMO

AIM: This study was designed to investigate the effect of the angiotensin II receptor blocker olmesartan alone, or in combination with standard treatment with a statin, pravastatin, on atherosclerosis development in APOE*3Leiden transgenic mice. METHODS AND RESULTS: Four groups of 15 mice received an atherogenic diet alone (plasma cholesterol 17.4 +/- 2.7 mmol/l) or supplemented with either 0.008% (w/w) olmesartan (9.3 mg/kg per day) (plasma cholesterol 16.4 +/- 3.9 mmol/l), 0.03% (w/w) pravastatin (35 mg/kg per day) (plasma cholesterol 14.6 +/- 2.6 mmol/l), or the combination of both (plasma cholesterol 14.5 +/- 2.9 mmol/l) for 6 months. Treatment with olmesartan or pravastatin reduced the development of atherosclerosis as compared to the control group (-46 and -39%, respectively). Pravastatin also reduced the severity of the lesions. As compared to control the combination of both treatments almost fully prevented atherosclerosis (-91%, P < 0.001) and strongly reduced lesion number (-69%), lesion severity (-79%), number of macrophages (-89%) and T lymphocytes (-86%) per cross-section. Treatment with olmesartan alone and in combination with pravastatin inhibited the adhesion of monocytes to the vessel wall (-22%; P < 0.05 and -25%; P < 0.01, respectively), and reduced the relative quantity of macrophages in the lesions (-38%; P < 0.05 and -26%; NS, respectively) as compared to control. CONCLUSION: Olmesartan reduced atherosclerosis development mainly by decreasing monocyte adhesion and the relative amount of macrophages, whereas pravastatin inhibited the progression of atherosclerosis to more advanced lesions, reflecting different anti-atherosclerotic modes of action of the two drugs. Combination therapy with olmesartan and pravastatin additively reduced atherosclerosis development, resulting in less and less severe lesions.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/administração & dosagem , Apolipoproteína E3/genética , Aterosclerose/prevenção & controle , Inibidores de Hidroximetilglutaril-CoA Redutases/administração & dosagem , Imidazóis/administração & dosagem , Pravastatina/administração & dosagem , Tetrazóis/administração & dosagem , Animais , Aterosclerose/sangue , Aterosclerose/genética , Aterosclerose/patologia , Pressão Sanguínea/efeitos dos fármacos , Colágeno/metabolismo , Dieta Aterogênica , Sinergismo Farmacológico , Feminino , Humanos , Inflamação/prevenção & controle , Lipídeos/sangue , Macrófagos/patologia , Camundongos , Camundongos Transgênicos , Monócitos/patologia , Músculo Liso Vascular/patologia , Proteínas Recombinantes/genética , Proteína Amiloide A Sérica/metabolismo , Linfócitos T/patologia
16.
Arterioscler Thromb Vasc Biol ; 26(11): 2560-6, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16931788

RESUMO

OBJECTIVE: We investigated whether the dual PPARalpha/gamma agonist tesaglitazar has anti-atherogenic effects in ApoE*3Leiden mice with reduced insulin sensitivity. METHODS AND RESULTS: ApoE*3Leiden transgenic mice were fed a high-fat (HF) insulin-resistance-inducing diet. One group received a high-cholesterol (HC) supplement (1% wt/wt; HC group). A second group received the same HC supplement along with tesaglitazar (T) 0.5 micromol/kg diet (T group). A third (control) group received a low-cholesterol (LC) supplement (0.1% wt/wt; LC group). Tesaglitazar decreased plasma cholesterol by 20% compared with the HC group; cholesterol levels were similar in the T and LC groups. Compared with the HC group, tesaglitazar caused a 92% reduction in atherosclerosis, whereas a 56% reduction was seen in the cholesterol-matched LC group. Furthermore, tesaglitazar treatment significantly reduced lesion number beyond that expected from cholesterol lowering and induced a shift to less severe lesions. Concomitantly, tesaglitazar reduced macrophage-rich and collagen areas. In addition, tesaglitazar reduced inflammatory markers, including plasma SAA levels, the number of adhering monocytes, and nuclear factor kappaB-activity in the vessel wall. CONCLUSIONS: Tesaglitazar has anti-atherosclerotic effects in the mouse model that go beyond plasma cholesterol lowering, possibly caused by a combination of altered lipoprotein profiles and anti-inflammatory vascular effects.


Assuntos
Alcanossulfonatos/farmacologia , Apolipoproteínas E/metabolismo , Aterosclerose/patologia , Hipercolesterolemia/fisiopatologia , Resistência à Insulina , PPAR alfa/agonistas , PPAR gama/agonistas , Fenilpropionatos/farmacologia , Animais , Apolipoproteína E3 , Aterosclerose/etiologia , Aterosclerose/fisiopatologia , Biomarcadores/sangue , Vasos Sanguíneos/patologia , Adesão Celular , Colesterol/sangue , Colesterol na Dieta/administração & dosagem , Colágeno/metabolismo , Gorduras na Dieta/administração & dosagem , Feminino , Hipercolesterolemia/sangue , Hipercolesterolemia/complicações , Inflamação/sangue , Lipídeos/sangue , Lipoproteínas/sangue , Macrófagos/patologia , Camundongos , Camundongos Transgênicos , Monócitos/patologia , NF-kappa B/metabolismo
17.
Sci Rep ; 7(1): 2915, 2017 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-28588299

RESUMO

Obesity-related albuminuria is associated with decline of kidney function and is considered a first sign of diabetic nephropathy. Suggested factors linking obesity to kidney dysfunction include low-grade inflammation, insulin resistance and adipokine dysregulation. Here, we investigated the effects of two pharmacological compounds with established anti-inflammatory properties, rosiglitazone and rosuvastatin, on kidney dysfunction during high-fat diet (HFD)-induced obesity. For this, human CRP transgenic mice were fed standard chow, a lard-based HFD, HFD+rosuvastatin or HFD+rosiglitazone for 42 weeks to study effects on insulin resistance; plasma inflammatory markers and adipokines; and renal pathology. Rosiglitazone but not rosuvastatin prevented HFD-induced albuminuria and renal fibrosis and inflammation. Also, rosiglitazone prevented HFD-induced KIM-1 expression, while levels were doubled with rosuvastatin. This was mirrored by miR-21 expression, which plays a role in fibrosis and is associated with renal dysfunction. Plasma insulin did not correlate with albuminuria. Only rosiglitazone increased circulating adiponectin concentrations. In all, HFD-induced albuminuria, and renal inflammation, injury and fibrosis is prevented by rosiglitazone but not by rosuvastatin. These beneficial effects of rosiglitazone are linked to lowered miR-21 expression but not connected with the selectively enhanced plasma adiponectin levels observed in rosiglitazone-treated animals.


Assuntos
Proteína C-Reativa/genética , Rim/efeitos dos fármacos , Rim/metabolismo , Substâncias Protetoras/farmacologia , Rosiglitazona/farmacologia , Adipocinas/sangue , Adiponectina/genética , Adiponectina/metabolismo , Animais , Biomarcadores , Glicemia , Dieta Hiperlipídica , Fibrose , Humanos , Imuno-Histoquímica , Mediadores da Inflamação/metabolismo , Insulina/sangue , Rim/patologia , Camundongos , Camundongos Transgênicos , MicroRNAs/genética
18.
PLoS One ; 12(7): e0180648, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28678821

RESUMO

BACKGROUND: Obesity frequently associates with the development of non-alcoholic fatty liver disease (NAFLD) and atherosclerosis. Chronic inflammation in white adipose tissue (WAT) seems to be an important driver of these manifestations. OBJECTIVE: This study investigated a combination of an extensively hydrolyzed casein (eHC), docosahexaenoic acid (DHA), arachidonic acid (ARA), and Lactobacillus Rhamnosus GG (LGG) (together referred to as nutritional ingredients, NI) on the development of obesity, metabolic risk factors, WAT inflammation, NAFLD and atherosclerosis in high-fat diet-fed LDLr-/-.Leiden mice, a model that mimics disease development in humans. METHODS: LDLr-/-.Leiden male mice (n = 15/group) received a high-fat diet (HFD, 45 Kcal%) for 21 weeks with or without the NI (23.7% eHC, 0.083% DHA, 0.166% ARA; all w/w and 1x109 CFU LGG gavage 3 times/week). HFD and HFD+NI diets were isocaloric. A low fat diet (LFD, 10 Kcal%) was used for reference. Body weight, food intake and metabolic risk factors were assessed over time. At week 21, tissues were analyzed for WAT inflammation (crown-like structures), NAFLD and atherosclerosis. Effects of the individual NI components were explored in a follow-up experiment (n = 7/group). RESULTS: When compared to HFD control, treatment with the NI strongly reduced body weight to levels of the LFD group, and significantly lowered (P<0.01) plasma insulin, cholesterol, triglycerides, leptin and serum amyloid A (P<0.01). NI also reduced WAT mass and inflammation. Strikingly, NI treatment significantly reduced macrovesicular steatosis, lobular inflammation and liver collagen (P<0.05), and attenuated atherosclerosis development (P<0.01). Of the individual components, the effects of eHC were most pronounced but could not explain the entire effects of the NI formulation. CONCLUSIONS: A combination of eHC, ARA, DHA and LGG attenuates obesity and associated cardiometabolic diseases (NAFLD, atherosclerosis) in LDLr-/-.Leiden mice. The observed reduction of inflammation in adipose tissue and in the liver provides a rationale for these comprehensive health effects.


Assuntos
Aterosclerose/prevenção & controle , Caseínas/administração & dosagem , Hepatopatia Gordurosa não Alcoólica/prevenção & controle , Obesidade/prevenção & controle , Receptores de LDL/fisiologia , Adiposidade/efeitos dos fármacos , Animais , Caseínas/farmacologia , Dieta Hiperlipídica , Masculino , Camundongos , Receptores de LDL/genética , Aumento de Peso
19.
Sci Rep ; 7: 43261, 2017 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-28256596

RESUMO

High-fat diets (HFD) are thought to contribute to the development of metabolism-related diseases. The long-term impact of HFD may be mediated by epigenetic mechanisms, and indeed, HFD has been reported to induce DNA methylation changes in white adipose tissue (WAT) near metabolism related genes. However, previous studies were limited to a single WAT depot, a single time-point and primarily examined the pre-pubertal period. To define dynamic DNA methylation patterns specific for WAT depots, we investigated DNA methylation of Pparg2 and Leptin in gonadal adipose tissue (GAT) and subcutaneous adipose tissue (SAT), at baseline and after 6, 12 and 24 weeks of HFD exposure in adult mice. HFD induced hypermethylation of both the Leptin promoter (max. 19.6% at week 24, P = 2.6·10-3) and the Pparg2 promoter in GAT (max. 10.5% at week 12, P = 0.001). The differential methylation was independent of immune cell infiltration upon HFD exposure. In contrast, no differential methylation in the Pparg2 and Leptin promoter was observed in SAT. Leptin and Pparg2 DNA methylation were correlated with gene expression in GAT. Our study shows that prolonged exposure to HFD in adulthood is associated with a gradually increasing DNA methylation level at the Leptin and Pparg2 promoters in a depot-specific manner.


Assuntos
Tecido Adiposo Branco/patologia , Metilação de DNA , Dieta Hiperlipídica , Epigênese Genética , Leptina/genética , PPAR gama/genética , Regiões Promotoras Genéticas , Animais , Gônadas/patologia , Camundongos Endogâmicos C57BL , Tela Subcutânea/patologia , Fatores de Tempo
20.
Hepatol Commun ; 1(4): 311-325, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-29404461

RESUMO

CAT-2003 is a novel conjugate of eicosapentaenoic acid (EPA) and niacin designed to be hydrolyzed by fatty acid amide hydrolase to release EPA inside cells at the endoplasmic reticulum. In cultured liver cells, CAT-2003 blocked the maturation of sterol regulatory element-binding protein (SREBP)-1 and SREBP-2 proteins and decreased the expression of multiple SREBP target genes, including HMGCR and PCSK9. Consistent with proprotein convertase subtilisin/kexin type 9 (PCSK9) reduction, both low-density lipoprotein receptor protein at the cell surface and low-density lipoprotein particle uptake were increased. In apolipoprotein E*3-Leiden mice fed a cholesterol-containing western diet, CAT-2003 decreased hepatic inflammation and steatosis as evidenced by fewer inflammatory cell aggregates in histopathologic sections, decreased nuclear factor kappa B activity in liver lysates, reduced inflammatory gene expression, reduced intrahepatic cholesteryl ester and triglyceride levels, and decreased liver mass. Plasma PCSK9 was reduced and hepatic low-density lipoprotein receptor protein expression was increased; plasma cholesterol and triglyceride levels were lowered. Aortic root segments showed reduction of several atherosclerotic markers, including lesion size, number, and severity. CAT-2003, when dosed in combination with atorvastatin, further lowered plasma cholesterol levels and decreased hepatic expression of SREBP target genes. Conclusion: SREBP inhibition is a promising new strategy for the prevention and treatment of diseases associated with abnormal lipid metabolism, such as atherosclerosis and nonalcoholic steatohepatitis. (Hepatology Communications 2017;1:311-325).

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA