Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Mol Syst Biol ; 16(4): e9495, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32337855

RESUMO

The prevalence of non-alcoholic fatty liver disease (NAFLD) continues to increase dramatically, and there is no approved medication for its treatment. Recently, we predicted the underlying molecular mechanisms involved in the progression of NAFLD using network analysis and identified metabolic cofactors that might be beneficial as supplements to decrease human liver fat. Here, we first assessed the tolerability of the combined metabolic cofactors including l-serine, N-acetyl-l-cysteine (NAC), nicotinamide riboside (NR), and l-carnitine by performing a 7-day rat toxicology study. Second, we performed a human calibration study by supplementing combined metabolic cofactors and a control study to study the kinetics of these metabolites in the plasma of healthy subjects with and without supplementation. We measured clinical parameters and observed no immediate side effects. Next, we generated plasma metabolomics and inflammatory protein markers data to reveal the acute changes associated with the supplementation of the metabolic cofactors. We also integrated metabolomics data using personalized genome-scale metabolic modeling and observed that such supplementation significantly affects the global human lipid, amino acid, and antioxidant metabolism. Finally, we predicted blood concentrations of these compounds during daily long-term supplementation by generating an ordinary differential equation model and liver concentrations of serine by generating a pharmacokinetic model and finally adjusted the doses of individual metabolic cofactors for future human clinical trials.


Assuntos
Acetilcisteína/administração & dosagem , Carnitina/administração & dosagem , Metabolômica/métodos , Niacinamida/análogos & derivados , Serina/administração & dosagem , Acetilcisteína/sangue , Adulto , Animais , Carnitina/sangue , Suplementos Nutricionais , Quimioterapia Combinada , Voluntários Saudáveis , Humanos , Masculino , Modelos Animais , Niacinamida/administração & dosagem , Niacinamida/sangue , Hepatopatia Gordurosa não Alcoólica/dietoterapia , Medicina de Precisão , Compostos de Piridínio , Ratos , Serina/sangue
2.
J Clin Endocrinol Metab ; 104(12): 6207-6219, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31369090

RESUMO

CONTEXT: Saturated fatty acid (SFA) vs polyunsaturated fatty acid (PUFA) may promote nonalcoholic fatty liver disease by yet unclear mechanisms. OBJECTIVE: To investigate if overeating SFA- and PUFA-enriched diets lead to differential liver fat accumulation in overweight and obese humans. DESIGN: Double-blind randomized trial (LIPOGAIN-2). Overfeeding SFA vs PUFA for 8 weeks, followed by 4 weeks of caloric restriction. SETTING: General community. PARTICIPANTS: Men and women who are overweight or have obesity (n = 61). INTERVENTION: Muffins, high in either palm (SFA) or sunflower oil (PUFA), were added to the habitual diet. MAIN OUTCOME MEASURES: Lean tissue mass (not reported here). Secondary and exploratory outcomes included liver and ectopic fat depots. RESULTS: By design, body weight gain was similar in SFA (2.31 ± 1.38 kg) and PUFA (2.01 ± 1.90 kg) groups, P = 0.50. SFA markedly induced liver fat content (50% relative increase) along with liver enzymes and atherogenic serum lipids. In contrast, despite similar weight gain, PUFA did not increase liver fat or liver enzymes or cause any adverse effects on blood lipids. SFA had no differential effect on the accumulation of visceral fat, pancreas fat, or total body fat compared with PUFA. SFA consistently increased, whereas PUFA reduced circulating ceramides, changes that were moderately associated with liver fat changes and proposed markers of hepatic lipogenesis. The adverse metabolic effects of SFA were reversed by calorie restriction. CONCLUSIONS: SFA markedly induces liver fat and serum ceramides, whereas dietary PUFA prevents liver fat accumulation and reduces ceramides and hyperlipidemia during excess energy intake and weight gain in overweight individuals.


Assuntos
Ceramidas/metabolismo , Gorduras na Dieta/efeitos adversos , Ácidos Graxos Insaturados/metabolismo , Fígado Gorduroso/etiologia , Hiperfagia/complicações , Obesidade/etiologia , Sobrepeso/etiologia , Adulto , Método Duplo-Cego , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Feminino , Seguimentos , Humanos , Lipídeos/análise , Masculino , Obesidade/metabolismo , Obesidade/patologia , Sobrepeso/metabolismo , Sobrepeso/patologia , Prognóstico , Aumento de Peso
3.
Arterioscler Thromb Vasc Biol ; 38(10): 2318-2326, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29903735

RESUMO

Objective- To investigate the effect of gut microbiota and diet on atherogenesis. Approach and Results- Here, we investigated the interaction between the gut microbiota and diet on atherosclerosis by feeding germ-free or conventionally raised Apoe-/- mice chow or Western diet alone or supplemented with choline (which is metabolized by the gut microbiota and host enzymes to trimethylamine N-oxide) for 12 weeks. We observed smaller aortic lesions and lower plasma cholesterol levels in conventionally raised mice compared with germ-free mice on a chow diet; these differences were not observed in mice on a Western diet. Choline supplementation increased plasma trimethylamine N-oxide levels in conventionally raised mice but not in germ-free mice. However, this treatment did not affect the size of aortic lesions or plasma cholesterol levels. Gut microbiota composition was analyzed by sequencing of 16S rRNA genes. As expected, the global community structure and relative abundance of many taxa differed between mice fed chow or a Western diet. Choline supplementation had minor effects on the community structure although the relative abundance of some taxa belonging to Clostridiales was altered. Conclusions- In conclusion, the impact of the gut microbiota on atherosclerosis is dietary dependent and is associated with plasma cholesterol levels. Furthermore, the microbiota was required for trimethylamine N-oxide production from dietary choline, but this process could not be linked to increased atherosclerosis in this model.


Assuntos
Doenças da Aorta/microbiologia , Aterosclerose/microbiologia , Bactérias/metabolismo , Colina/administração & dosagem , Dieta Ocidental , Suplementos Nutricionais , Microbioma Gastrointestinal , Intestinos/microbiologia , Camundongos Knockout para ApoE , Ração Animal , Animais , Doenças da Aorta/sangue , Doenças da Aorta/genética , Doenças da Aorta/prevenção & controle , Aterosclerose/sangue , Aterosclerose/genética , Aterosclerose/prevenção & controle , Bactérias/genética , Bactérias/crescimento & desenvolvimento , Colesterol/sangue , Colina/metabolismo , Modelos Animais de Doenças , Masculino , Metilaminas/metabolismo , Camundongos Endogâmicos C57BL , Ribotipagem
4.
PLoS Genet ; 13(9): e1007004, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28886012

RESUMO

Dietary fatty acids can be incorporated directly into phospholipids. This poses a specific challenge to cellular membranes since their composition, hence properties, could greatly vary with different diets. That vast variations in diets are tolerated therefore implies the existence of regulatory mechanisms that monitor and regulate membrane compositions. Here we show that the adiponectin receptor AdipoR2, and its C. elegans homolog PAQR-2, are essential to counter the membrane rigidifying effects of exogenously provided saturated fatty acids. In particular, we use dietary supplements or mutated E. coli as food, together with direct measurements of membrane fluidity and composition, to show that diets containing a high ratio of saturated to monounsaturated fatty acids cause membrane rigidity and lethality in the paqr-2 mutant. We also show that mammalian cells in which AdipoR2 has been knocked-down by siRNA are unable to prevent the membrane-rigidifying effects of palmitic acid. We conclude that the PAQR-2 and AdipoR2 proteins share an evolutionarily conserved function that maintains membrane fluidity in the presence of exogenous saturated fatty acids.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Membrana Celular/genética , Fluidez de Membrana/genética , Proteínas de Membrana/genética , Receptores de Adiponectina/genética , Animais , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Membrana Celular/química , Membrana Celular/metabolismo , Ácidos Graxos/química , Ácidos Graxos/metabolismo , Células HEK293 , Humanos , Proteínas de Membrana/metabolismo , Fosfolipídeos/química , Fosfolipídeos/genética , RNA Interferente Pequeno , Receptores de Adiponectina/metabolismo
5.
Mol Syst Biol ; 13(3): 916, 2017 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-28254760

RESUMO

To elucidate the molecular mechanisms underlying non-alcoholic fatty liver disease (NAFLD), we recruited 86 subjects with varying degrees of hepatic steatosis (HS). We obtained experimental data on lipoprotein fluxes and used these individual measurements as personalized constraints of a hepatocyte genome-scale metabolic model to investigate metabolic differences in liver, taking into account its interactions with other tissues. Our systems level analysis predicted an altered demand for NAD+ and glutathione (GSH) in subjects with high HS Our analysis and metabolomic measurements showed that plasma levels of glycine, serine, and associated metabolites are negatively correlated with HS, suggesting that these GSH metabolism precursors might be limiting. Quantification of the hepatic expression levels of the associated enzymes further pointed to altered de novo GSH synthesis. To assess the effect of GSH and NAD+ repletion on the development of NAFLD, we added precursors for GSH and NAD+ biosynthesis to the Western diet and demonstrated that supplementation prevents HS in mice. In a proof-of-concept human study, we found improved liver function and decreased HS after supplementation with serine (a precursor to glycine) and hereby propose a strategy for NAFLD treatment.


Assuntos
Glutationa/metabolismo , Lipoproteínas/metabolismo , Metabolômica/métodos , NAD/metabolismo , Hepatopatia Gordurosa não Alcoólica/genética , Serina/administração & dosagem , Animais , Modelos Animais de Doenças , Feminino , Regulação Enzimológica da Expressão Gênica , Genoma , Glicina/sangue , Humanos , Fígado/enzimologia , Fígado/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Hepatopatia Gordurosa não Alcoólica/dietoterapia , Hepatopatia Gordurosa não Alcoólica/metabolismo , Modelagem Computacional Específica para o Paciente , Serina/sangue , Serina/uso terapêutico
6.
J Nutr Biochem ; 33: 111-8, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27155917

RESUMO

Studies on metabolism of polyphenols have revealed extensive transformations in the carbon backbone by colonic microbiota; however, the influence of microbial and hepatic transformations on human urinary metabolites has not been explored. Therefore, the aims of this study were (1) to compare the in vitro microbial phenolic metabolite profile of foods and beverages with that excreted in urine of subjects consuming the same foodstuff and (2) to explore the role of liver on postcolonic metabolism of polyphenols by using in vitro hepatic models. A 24-h urinary phenolic metabolite profile was evaluated in 72 subjects participating in an 8-week clinical trial during which they were randomly assigned to diets differing for polyphenol content. Polyphenol-rich foods and beverages used in the clinical trial were subjected to human fecal microbiota in the in vitro colon model. Metabolites from green tea, one of the main components of the polyphenol-rich diet, were incubated with primary hepatocytes to highlight hepatic conversion of polyphenols. The analyses were performed using targeted gas chromatography with mass spectrometer (GCxGC-TOFMS:colon model; GC-MS: urine and hepatocytes). A significant correlation was found between urinary and colonic metabolites with C1-C3 side chain (P=.040). However, considerably higher amounts of hippuric acid, 3-hydroxybenzoic acid and ferulic acid were detected in urine than in the colon model. The hepatic conversion showed additional amounts of these metabolites complementing the gap between in vitro colon model and the in vivo urinary excretion. Therefore, combining in vitro colon and hepatic models may better elucidate the metabolism of polyphenols from dietary exposure to urinary metabolites.


Assuntos
Colo/microbiologia , Dieta , Microbioma Gastrointestinal , Fígado/metabolismo , Modelos Biológicos , Sobrepeso/metabolismo , Polifenóis/metabolismo , Adulto , Algoritmos , Células Cultivadas , Ácidos Cumáricos/metabolismo , Ácidos Cumáricos/urina , Fezes/microbiologia , Manipulação de Alimentos , Hipuratos/metabolismo , Hipuratos/urina , Humanos , Hidroxibenzoatos/metabolismo , Hidroxibenzoatos/urina , Mucosa Intestinal/microbiologia , Fígado/citologia , Obesidade/metabolismo , Obesidade/urina , Sobrepeso/urina , Oxirredução , Polifenóis/administração & dosagem , Polifenóis/urina , Chá/química
7.
Infect Immun ; 84(4): 1205-1213, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26857576

RESUMO

Sepsis caused by Staphylococcus aureus is increasing in incidence. With the alarming use of antibiotics,S. aureus is prone to become methicillin resistant. Antibiotics are the only widely used pharmacological treatment for sepsis. Interestingly, mice fed high-fat diet (HFD) rich in polyunsaturated fatty acids have better survival of S. aureus-induced sepsis than mice fed HFD rich in saturated fatty acids (HFD-S). To investigate what component of polyunsaturated fatty acids, i.e., omega-3 or omega-6 fatty acids, exerts beneficial effects on the survival of S. aureus-induced sepsis, mice were fed HFD rich in omega-3 or omega-6 fatty acids for 8 weeks prior to inoculation with S. aureus Further, mice fed HFD-S were treated with omega-3 fatty acid metabolites known as resolvins. Mice fed HFD rich in omega-3 fatty acids had increased survival and decreased bacterial loads compared to those for mice fed HFD-S after S. aureus-induced sepsis. Furthermore, the bacterial load was decreased in resolvin-treated mice fed HFD-S after S. aureus-induced sepsis compared with that in mice treated with vehicle. Dietary omega-3 fatty acids increase the survival of S. aureus-induced sepsis by reversing the deleterious effect of HFD-S on mouse survival.


Assuntos
Carga Bacteriana/efeitos dos fármacos , Ácidos Graxos Ômega-3/farmacologia , Sepse/microbiologia , Infecções Estafilocócicas/dietoterapia , Staphylococcus aureus , Tecido Adiposo , Animais , Citocinas/genética , Citocinas/metabolismo , Ácidos Graxos/administração & dosagem , Ácidos Graxos/farmacologia , Ácidos Graxos Ômega-3/administração & dosagem , Inflamação/metabolismo , Camundongos , Distribuição Aleatória , Sepse/dietoterapia , Infecções Estafilocócicas/microbiologia
8.
PLoS One ; 10(6): e0130898, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26098110

RESUMO

OBJECTIVE: The first objective was to investigate if intracellular and extracellular levels of reactive oxygen species (ROS) within the mouse aorta increase before or after diet-induced lesion formation. The second objective was to investigate if intracellular and extracellular ROS correlates to cell composition in atherosclerotic lesions. The third objective was to investigate if intracellular and extracellular ROS levels within established atherosclerotic lesions can be reduced by lipid lowering by diet or atorvastatin. APPROACH AND RESULTS: To address our objectives, we established a new imaging technique to visualize and quantify intracellular and extracellular ROS levels within intact mouse aortas ex vivo. Using this technique, we found that intracellular, but not extracellular, ROS levels increased prior to lesion formation in mouse aortas. Both intracellular and extracellular ROS levels were increased in advanced lesions. Intracellular ROS correlated with lesion content of macrophages. Extracellular ROS correlated with lesion content of smooth muscle cells. The high levels of ROS in advanced lesions were reduced by 5 days high dose atorvastatin treatment but not by lipid lowering by diet. Atorvastatin treatment did not affect lesion inflammation (aortic arch mRNA levels of CXCL 1, ICAM-1, MCP-1, TNF-α, VCAM, IL-6, and IL-1ß) or cellular composition (smooth muscle cell, macrophage, and T-cell content). CONCLUSIONS: Aortic levels of intracellular ROS increase prior to lesion formation and may be important in initiation of atherosclerosis. Our results suggest that within lesions, macrophages produce mainly intracellular ROS whereas smooth muscle cells produce extracellular ROS. Short term atorvastatin treatment, but not lipid lowering by diet, decreases ROS levels within established advanced lesions; this may help explain the lesion stabilizing and anti-inflammatory effects of long term statin treatment.


Assuntos
Aorta/metabolismo , Atorvastatina/farmacologia , Doença da Artéria Coronariana/patologia , Espécies Reativas de Oxigênio/metabolismo , Análise de Variância , Animais , Aorta/patologia , Apolipoproteínas E/genética , Benzimidazóis , Dieta com Restrição de Gorduras , Feminino , Lipídeos/sangue , Medições Luminescentes , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Eur Heart J ; 36(36): 2425-37, 2015 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-26009596

RESUMO

Familial hypercholesterolaemia (FH) is a common genetic cause of premature coronary heart disease (CHD). Globally, one baby is born with FH every minute. If diagnosed and treated early in childhood, individuals with FH can have normal life expectancy. This consensus paper aims to improve awareness of the need for early detection and management of FH children. Familial hypercholesterolaemia is diagnosed either on phenotypic criteria, i.e. an elevated low-density lipoprotein cholesterol (LDL-C) level plus a family history of elevated LDL-C, premature coronary artery disease and/or genetic diagnosis, or positive genetic testing. Childhood is the optimal period for discrimination between FH and non-FH using LDL-C screening. An LDL-C ≥5 mmol/L (190 mg/dL), or an LDL-C ≥4 mmol/L (160 mg/dL) with family history of premature CHD and/or high baseline cholesterol in one parent, make the phenotypic diagnosis. If a parent has a genetic defect, the LDL-C cut-off for the child is ≥3.5 mmol/L (130 mg/dL). We recommend cascade screening of families using a combined phenotypic and genotypic strategy. In children, testing is recommended from age 5 years, or earlier if homozygous FH is suspected. A healthy lifestyle and statin treatment (from age 8 to 10 years) are the cornerstones of management of heterozygous FH. Target LDL-C is <3.5 mmol/L (130 mg/dL) if >10 years, or ideally 50% reduction from baseline if 8-10 years, especially with very high LDL-C, elevated lipoprotein(a), a family history of premature CHD or other cardiovascular risk factors, balanced against the long-term risk of treatment side effects. Identifying FH early and optimally lowering LDL-C over the lifespan reduces cumulative LDL-C burden and offers health and socioeconomic benefits. To drive policy change for timely detection and management, we call for further studies in the young. Increased awareness, early identification, and optimal treatment from childhood are critical to adding decades of healthy life for children and adolescents with FH.


Assuntos
Hiperlipoproteinemia Tipo II/tratamento farmacológico , Adolescente , Adulto , Aterosclerose/diagnóstico , Aterosclerose/tratamento farmacológico , Espessura Intima-Media Carotídea , Criança , Técnicas de Laboratório Clínico/métodos , Efeitos Psicossociais da Doença , Aconselhamento , Dieta , Suplementos Nutricionais , Diagnóstico Precoce , Economia Médica , Medicina Baseada em Evidências , Feminino , Testes Genéticos , Heterozigoto , Homozigoto , Humanos , Hiperlipoproteinemia Tipo II/diagnóstico , Hiperlipoproteinemia Tipo II/genética , Expectativa de Vida , Adesão à Medicação , Pessoa de Meia-Idade , Gravidez , Complicações na Gravidez/etiologia , Fatores de Risco , Adulto Jovem
10.
Eur Heart J ; 32(11): 1345-61, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21531743

RESUMO

Even at low-density lipoprotein cholesterol (LDL-C) goal, patients with cardiometabolic abnormalities remain at high risk of cardiovascular events. This paper aims (i) to critically appraise evidence for elevated levels of triglyceride-rich lipoproteins (TRLs) and low levels of high-density lipoprotein cholesterol (HDL-C) as cardiovascular risk factors, and (ii) to advise on therapeutic strategies for management. Current evidence supports a causal association between elevated TRL and their remnants, low HDL-C, and cardiovascular risk. This interpretation is based on mechanistic and genetic studies for TRL and remnants, together with the epidemiological data suggestive of the association for circulating triglycerides and cardiovascular disease. For HDL, epidemiological, mechanistic, and clinical intervention data are consistent with the view that low HDL-C contributes to elevated cardiovascular risk; genetic evidence is unclear however, potentially reflecting the complexity of HDL metabolism. The Panel believes that therapeutic targeting of elevated triglycerides (≥ 1.7 mmol/L or 150 mg/dL), a marker of TRL and their remnants, and/or low HDL-C (<1.0 mmol/L or 40 mg/dL) may provide further benefit. The first step should be lifestyle interventions together with consideration of compliance with pharmacotherapy and secondary causes of dyslipidaemia. If inadequately corrected, adding niacin or a fibrate, or intensifying LDL-C lowering therapy may be considered. Treatment decisions regarding statin combination therapy should take into account relevant safety concerns, i.e. the risk of elevation of blood glucose, uric acid or liver enzymes with niacin, and myopathy, increased serum creatinine and cholelithiasis with fibrates. These recommendations will facilitate reduction in the substantial cardiovascular risk that persists in patients with cardiometabolic abnormalities at LDL-C goal.


Assuntos
Doenças Cardiovasculares/etiologia , HDL-Colesterol/metabolismo , Dislipidemias/complicações , Lipoproteínas/metabolismo , Triglicerídeos/metabolismo , Doenças Cardiovasculares/sangue , Doenças Cardiovasculares/prevenção & controle , Ensaios Clínicos como Assunto , Dislipidemias/sangue , Dislipidemias/prevenção & controle , Ácidos Graxos Ômega-3/uso terapêutico , Ácidos Fíbricos/uso terapêutico , Previsões , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Hipolipemiantes/uso terapêutico , Estilo de Vida , Metabolismo dos Lipídeos , Niacina/uso terapêutico , Fatores de Risco
11.
J Lipid Res ; 47(1): 67-77, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16227197

RESUMO

Epigallocatechin gallate (EGCG) increases the formation of cytosolic lipid droplets by a mechanism that is independent of the rate of triglyceride biosynthesis and involves an enhanced fusion between lipid droplets, a process that is crucial for their growth in size. EGCG treatment reduced the secretion of both triglycerides and apolipoprotein B-100 (apoB-100) VLDLs but not of transferrin, albumin, or total proteins, indicating that EGCG diverts triglycerides from VLDL assembly to storage in the cytosol. This is further supported by the observed increase in both intracellular degradation of apoB-100 and ubiquitination of the protein (indicative of increased proteasomal degradation) in EGCG-treated cells. EGCG did not interfere with the microsomal triglyceride transfer protein, and the effect of EGCG on the secretion of VLDLs was found to be independent of the LDL receptor. Thus, our results indicate that EGCG promotes the accumulation of triglycerides in cytosolic lipid droplets, thereby diverting lipids from the assembly of VLDL to storage in the cytosol. Our results also indicate that the accumulation of lipids in the cytosol is not always associated with increased secretion of VLDL.


Assuntos
Apolipoproteínas B/metabolismo , Catequina/análogos & derivados , Metabolismo dos Lipídeos/efeitos dos fármacos , Lipoproteínas VLDL/metabolismo , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Animais , Apolipoproteína B-100 , Apolipoproteínas B/genética , Sequência de Bases , Catequina/farmacologia , Linhagem Celular , Citosol/efeitos dos fármacos , Citosol/metabolismo , DNA Complementar/genética , Heparina/farmacologia , Humanos , Lipídeos/sangue , Lipoproteínas/sangue , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Células NIH 3T3 , Perilipina-2 , Ratos , Receptores de LDL/efeitos dos fármacos , Receptores de LDL/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA