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











Intervalo de año de publicación
1.
Fundam Clin Pharmacol ; : e13024, 2024 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-38923017

RESUMEN

BACKGROUND: Monotherapy to treat obesity-associated liver insult is limited. OBJECTIVES: In diet-induced obese mice showing metabolic dysfunction-associated steatotic liver disease (MASLD), we aimed to compare the combinations of sodium-glucose cotransporter-2 inhibitor (SGLT2i, empagliflozin, E), dipeptidyl peptidase-4 inhibitor (DPP4i, linagliptin, L), and glucagon-like peptide type 1 receptor agonist (GLP1RA, dulaglutide, D). METHODS: Male 3-month-old C57BL/6J mice were fed for 12 weeks in a control (C, n = 10) or high-fat (HF, n = 30) diet. Then, mice were followed for three additional weeks: C, HF, HF E + L, and HF E + D (n = 10/group). RESULTS: HF versus C showed higher hepatic triacylglycerol (TAG, +82%), steatosis (+850%), glucose intolerance (+71%), insulin (+98%), and insulin resistance (+68%). Compared to the HF group, HF E + L showed lower glucose intolerance (-60%), insulin (-61%), insulin resistance (-46%), TAG (-61%), and steatosis (-58%), and HF E + D showed lower glucose intolerance (-71%), insulin (-58%), insulin resistance (-62%), TAG (-61%), and steatosis (-82%). The principal component analysis (PCA) placed the HF group and the HF E + D group on opposite sides, while the HF E + L group was placed between C and HF E + D. CONCLUSION: PCA separated the groups considering the metabolism-related genes (glucose and lipid), mitochondrial biogenesis, and steatosis. The two pharmacological combinations showed beneficial effects in treating obesity and MASLD. However, the combination of SGLT2i and GLP1RA showed more potent beneficial effects on MASLD than SGLT2i and DPP4i and, therefore, should be the recommended combination.

2.
Front Physiol ; 14: 1145233, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37064892

RESUMEN

Chronic kidney disease (CKD) has been recognized as a significant global health problem due to being an important contributor to morbidity and mortality. Inflammation is the critical event that leads to CKD development orchestrated by a complex interaction between renal parenchyma and immune cells. Particularly, the crosstalk between tubular epithelial cells (TECs) and macrophages is an example of the critical cell communication in the kidney that drives kidney fibrosis, a pathological feature in CKD. Metabolism dysregulation of TECs and macrophages can be a bridge that connects inflammation and fibrogenesis. Currently, some evidence has reported how cellular lipid disturbances can affect kidney disease and cause tubulointerstitial fibrosis highlighting the importance of investigating potential molecules that can restore metabolic parameters. Vitamin D (VitD) is a hormone naturally produced by mammalian cells in a coordinated manner by the skin, liver, and kidneys. VitD deficiency or insufficiency is prevalent in patients with CKD, and serum levels of VitD are inversely correlated with the degree of kidney inflammation and renal function. Proximal TECs and macrophages produce the active form of VitD, and both express the VitD receptor (VDR) that evidence the importance of this nutrient in regulating their functions. However, whether VitD signaling drives physiological and metabolism improvement of TECs and macrophages during kidney injury is an open issue to be debated. In this review, we brought to light VitD as an important metabolic modulator of lipid metabolism in TECs and macrophages. New scientific approaches targeting VitD e VDR signaling at the cellular metabolic level can provide a better comprehension of its role in renal physiology and CKD progression.

3.
J Clin Med ; 12(4)2023 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-36836104

RESUMEN

BACKGROUND: Pulmonary artery hypertension (PAH) is a chronic and progressive disease. Although current therapy has improved the disease prognosis, PAH has a poor survival rate. The key feature leading to disease progression and death is right ventricular (RV) failure. METHODS AND RESULTS: We assessed the role of trimetazidine, a fatty acid beta-oxidation (FAO) inhibitor, in right ventricular function, remodeling, and functional class in PAH patients, with a placebo-controlled double-blind, case-crossover trial. Twenty-seven PAH subjects were enrolled, randomized, and assigned to trimetazidine or placebo for three months and then reallocated to the other study arm. The primary endpoint was RV morphology and function change after three months of treatment. Secondary endpoints were the change in exercise capacity assessed by a 6 min walk test after three months of treatment and the change in pro-BNP and Galectin-3 plasma levels after three months. Trimetazidine use was safe and well-tolerated. After three months of treatment, patients in the trimetazidine group showed a small but significant reduction of RV diastolic area, and a substantial increase in the 6 min walk distance (418 vs. 438 mt, p = 0.023), without significant changes in biomarkers. CONCLUSIONS: A short course of trimetazidine is safe and well-tolerated on PAH patients, and it is associated with significant increases in the 6MWT and minor but significant improvement in RV remodeling. The therapeutic potential of this drug should be evaluated in larger clinical trials.

4.
São Paulo; s.n; s.n; 2023. 52 p. tab, graf, ilus.
Tesis en Portugués | LILACS | ID: biblio-1437572

RESUMEN

A obesidade está associada ao desenvolvimento de doenças crônicas não transmissíveis como hipertensão, resistência insulínica, dislipidemia e esteatose hepática. O consumo de compostos bioativos impacta na manutenção da saúde e na prevenção de risco de desenvolvimento dessas doenças. Entre os compostos bioativos, os monoterpenos são pouco investigados, apesar da literatura demonstrar efeitos promissores desses compostos sobre o metabolismo. O D-limoneno, o principal monoterpeno encontrado na laranja, é caracterizado por possuir efeitos hipolipemiantes, anti-inflamatórios e anti-obesogênicos. Estudos in vitro e in vivo descrevem sua capacidade de promover a ß-oxidação de ácidos graxos em adipócitos e redução da inflamação. Este estudo teve como objetivo investigar o efeito do D-limoneno no metabolismo e inflamação em um modelo de obesidade induzida por dieta. Para isso, quarenta camundongos machos (C57/Bl6) de 11 semanas de idade, foram distribuídos em 4 grupos, sendo que um dos grupos recebeu ração normolipídica e os demais, ração hiperlipídica. O D-limoneno foi suplementado na ração de dois grupos que receberam dieta hiperlipídica nas concentrações de 0,1%, e 0,8%. Considerando-se a ingestão alimentar dos animais, a ração suplementada com 0,1% D-limoneno correspondeu à ingestão de 0,15 g/kg/dia e ração com 0,8% de D-limoneno correspondeu a 1,3 g/kg/dia. Os animais tiveram o peso e a ingestão alimentar monitorados ao longo da intervenção com duração de 7 semanas. Os camundongos que receberam D-limoneno a 0,1% apresentaram menor ganho de peso e de acúmulo de tecido adiposo, comparado com os animais sem suplementação alimentados com a dieta hiperlipídica. Além disso, o D-limoneno promoveu a diminuição da concentração plasmática de marcadores inflamatórios incluindo TNF-α, INF-γ e IL-6 nos animais dos grupos que foram suplementados com D-limoneno. Entretanto, não houve diferença nos marcadores bioquímicos e metabólicos. Uma limitação do estudo foi o fato das complicações metabólicas associadas ao modelo de obesidade não terem sido plenamente estabelecidas, dados o alojamento individual, à curta duração da exposição à ração hiperlipídica e idade dos animais no início da suplementação. Esse fato pode ter dificultado a observação dos efeitos do D-limoneno na reversão dos parâmetros que seriam normalmente deteriorados pelo desenvolvimento da obesidade. Concluímos que o D-limoneno pode interferir no metabolismo energético, com possível efeito anti-obesogênico e anti-inflamatório. Devido às limitações do modelo, são necessários mais estudos para confirmar esses resultados


Obesity is associated with the development of chronic non-communicable diseases such as hypertension, insulin resistance, dyslipidemia, and hepatic steatosis. The intake of dietary bioactive compounds is associated with the maintenance of health and the prevention of chronic diseases. Among the group of bioactive compounds, monoterpenes are poorly investigated, in spite of several reports of their promising effects on metabolism. D-limonene is the main monoterpene found in oranges, known for its hypolipemic, anti-inflammatory, and anti-obesogenic effects. in vitro and in vivo studies associate D-limonene to increased ß-oxidation of fatty acids in adipocytes and reduced inflammation. This study aimed at investigating the effects of D-limonene on metabolism and inflammation in a diet-induced obesity model. For this purpose, forty male mice (C57/Bl6) were distributed in 4 groups, with one group receiving a normolipidic diet and the others, a high-fat diet. D-limonene was supplemented in the diets of two groups that received high-fat diet at the concentrations of 0.1% and 0.8%. Considering the feed intake, mice receiving D-limonene supplementation at 0.1% ingested in average 0.15 g/kg/day, while the mice receiving the supplemmentation at 0.8%, ingested approximately 1.3 g of D-limonene /kg/day. The animals had their weight and food intake monitored throughout the intervention. Mice that received Dlimonene supplementation at 0.1% showed reduced weight gain and accumulation of adipose tissue compared to the non-supplemented mice fed the high-fat diet. In addition, D-limonene promoted a decrease in hepatic inflammatory markers including TNF-α, INF-γ, and IL-6. However, there was no difference in biochemical and metabolic markers. A limitation of the study was that the metabolic complications associated with the obesity model were not fully established, probably due to the age at the start of the protocol (11 weeks), individual housing and short duration of the exposure to the high-fat feed. This fact may have prevented the observation of the positive effects of D-limonene in reversing parameters that would normally be impaired by the development of obesity. We conclude that D-limonene may interfere in energy metabolism, with a possible anti-obesogenic and anti-inflammatory effect. Due to the limitations of the model, further studies are needed to confirm these findings


Asunto(s)
Animales , Masculino , Ratones , Limoneno/efectos adversos , Obesidad/inducido químicamente , Técnicas In Vitro/métodos , Enfermedad Crónica/clasificación , Citrus sinensis/metabolismo , Monoterpenos/análisis , Dieta Alta en Grasa/efectos adversos , Inflamación/complicaciones , Antiinflamatorios/administración & dosificación
5.
Mol Cell Endocrinol ; 509: 110804, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32259637

RESUMEN

AIM: To investigate the effects of linagliptin treatment on hepatic energy metabolism and ER stress in high-fat-fed C57BL/6 mice. METHODS: Forty male C57BL/6 mice, three months of age, received a control diet (C, 10% of lipids as energy, n = 20) or high-fat diet (HF, 50% of lipids as energy, n = 20) for 10 weeks. The groups were randomly subdivided into four groups to receive linagliptin, for five weeks, at a dose of 30 mg/kg/day added to the diets: C, C-L, HF, and HF-L groups. RESULTS: The HF group showed higher body mass, total and hepatic cholesterol levels and total and hepatic triacylglycerol levels than the C group, all of which were significantly diminished by linagliptin in the HF-L group. The HF group had higher hepatic steatosis than the C group, whereas linagliptin markedly reduced the hepatic steatosis (less 52%, P < 0.001). The expression of Sirt1 and Pgc1a was more significant in the HF-L group than in the HF group. Linagliptin also elicited enhanced GLP-1 concentrations and a reduction in the expression of the lipogenic genes Fas and Srebp1c. Besides, HF-L showed a reduction in the genes related to endoplasmic reticulum stress Chop, Atf4, and Gadd45 coupled with reduced apoptotic nuclei immunostaining. CONCLUSION: Linagliptin caused a marked reduction in hepatic steatosis as a secondary effect of its glucose-lowering property. NAFLD countering involved reduced lipogenesis, increased beta-oxidation, and relief in endoplasmic reticulum stress, leading to reduced apoptosis and better preservation of the hepatic structure. Therefore, linagliptin may be used, preferably in diabetic patients, to avoid the progression of hepatic steatosis.


Asunto(s)
Dieta Alta en Grasa , Estrés del Retículo Endoplásmico , Conducta Alimentaria , Linagliptina/uso terapéutico , Lipogénesis , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Biomarcadores/metabolismo , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , Metabolismo de los Hidratos de Carbono/efectos de los fármacos , Ingestión de Alimentos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Ayuno/sangre , Péptido 1 Similar al Glucagón/sangre , Insulina/sangre , Resistencia a la Insulina , Linagliptina/farmacología , Gotas Lipídicas/efectos de los fármacos , Gotas Lipídicas/metabolismo , Lípidos/sangre , Lipogénesis/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/enzimología , Hígado/patología , Masculino , Ratones Endogámicos C57BL , Oxidación-Reducción , Perilipina-2/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
6.
Mol Cell Endocrinol ; 474: 227-237, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-29580823

RESUMEN

Endoplasmic reticulum (ER) stress and hepatic steatosis are intertwined with insulin resistance. PPARs are at the crossroads of these pathways. This study aimed to investigate the effects of GW0742 (PPAR-beta agonist) on hepatic energy metabolism and ER stress in a murine diet-induced obesity model. HF diet caused overweight, hyperinsulinemia, hepatic inflammation (increased NF-kB, TNF-alpha, and IL-6 protein expression) and favored hepatic lipogenesis, leading to ER stress, with ultrastructural and molecular alterations, ending up in proapoptotic stimulus. GW0742 rescued the overweight and the glucose tolerance, tackled hepatic inflammation and favored hepatic beta-oxidation over lipogenesis. These results comply with ER ultrastructure improvement, reducing ER stress and apoptosis in treated animals. Our results indicate that the PPAR-beta/delta activation alleviated the ER stress by improving the insulin sensitivity and maximizing the hepatic energy metabolism with a shift towards beta-oxidation. PPAR-beta/delta activation could be an essential tool to avoid the NAFLD progression and other obesity constraints.


Asunto(s)
Estrés del Retículo Endoplásmico/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Conducta Alimentaria , Hígado/metabolismo , Hígado/patología , PPAR-beta/agonistas , Tiazoles/farmacología , Alanina Transaminasa/metabolismo , Animales , Apoptosis/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Colesterol/metabolismo , Dieta Alta en Grasa , Ingestión de Energía/efectos de los fármacos , Hígado Graso/metabolismo , Hígado Graso/patología , Prueba de Tolerancia a la Glucosa , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/ultraestructura , Inflamación/patología , Resistencia a la Insulina , Lipogénesis/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/ultraestructura , Masculino , Ratones Endogámicos C57BL , Oxidación-Reducción/efectos de los fármacos , Isoformas de Proteínas/metabolismo
7.
Lipids Health Dis ; 17(1): 55, 2018 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-29554895

RESUMEN

BACKGROUND: We have recently demonstrated that palmitoleic acid (16:1n7) increases lipolysis, glucose uptake and glucose utilization for energy production in white adipose cells. In the present study, we tested the hypothesis that palmitoleic acid modulates bioenergetic activity in white adipocytes. METHODS: For this, 3 T3-L1 pre-adipocytes were differentiated into mature adipocytes in the presence (or absence) of palmitic (16:0) or palmitoleic (16:1n7) acid at 100 or 200 µM. The following parameters were evaluated: lipolysis, lipogenesis, fatty acid (FA) oxidation, ATP content, oxygen consumption, mitochondrial mass, citrate synthase activity and protein content of mitochondrial oxidative phosphorylation (OXPHOS) complexes. RESULTS: Treatment with 16:1n7 during 9 days raised basal and isoproterenol-stimulated lipolysis, FA incorporation into triacylglycerol (TAG), FA oxidation, oxygen consumption, protein expression of subunits representing OXPHOS complex II, III, and V and intracellular ATP content. These effects were not observed in adipocytes treated with 16:0. CONCLUSIONS: Palmitoleic acid, by concerted action on lipolysis, FA esterification, mitochondrial FA oxidation, oxygen consumption and ATP content, does enhance white adipocyte energy expenditure and may act as local hormone.


Asunto(s)
Adenosina Trifosfato/metabolismo , Adipocitos Blancos/efectos de los fármacos , Adipocitos Blancos/metabolismo , Ácidos Grasos Monoinsaturados/farmacología , Ácidos Grasos/metabolismo , Consumo de Oxígeno/efectos de los fármacos , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Animales , Metabolismo de los Lípidos/efectos de los fármacos , Lipogénesis/efectos de los fármacos , Lipólisis/efectos de los fármacos , Ratones , Oxidación-Reducción/efectos de los fármacos , Triglicéridos/metabolismo
8.
Cell Biochem Funct ; 35(8): 527-537, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29243276

RESUMEN

The purpose of this study was to determine if Shc proteins influence the metabolic response to acute (7 days) feeding of a high-fat diet (HFD). To this end, whole animal energy expenditure (EE) and substrate oxidation were measured in the Shc knockout (ShcKO) and wild-type (WT) mice fed a control or HFD. The activities of enzymes of glycolysis, the citric acid cycle, electron transport chain (ETC), and ß-oxidation were also investigated in liver and skeletal muscle of ShcKO and WT animals. The study showed that ShcKO increases (P < .05) EE adjusted for either total body weight or lean mass. This change in EE could contribute to decreases in weight gain in ShcKO versus WT mice fed an HFD. Thus, our results indicate that Shc proteins should be considered as potential targets for developing interventions to mitigate weight gain on HFD by stimulating EE. Although decreased levels of Shc proteins influenced the activity of some enzymes in response to high-fat feeding (eg, increasing the activity of acyl-CoA dehydrogenase), it did not produce concerted changes in enzymes of glycolysis, citric acid cycle, or the ETC. The physiological significance of observed changes in select enzyme activities remains to be determined. SIGNIFICANCE OF THE STUDY: We report higher EE in ShcKO versus WT mice when consuming the HFD. Although decreased levels of Shc proteins influenced the activity of a central enzyme of ß-oxidation in response to high-fat feeding, it did not produce concerted changes in enzymes of glycolysis, citric acid cycle, or the ETC. Thus, an increase in EE in response to consumption of an HFD may be a mechanism that leads to decreased weight gain previously reported in ShcKO mice with long-term consumption of an HFD.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Metabolismo Energético , Proteínas Adaptadoras de la Señalización Shc/metabolismo , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Adaptadoras de la Señalización Shc/deficiencia , Aumento de Peso
9.
Anim. Reprod. ; 14(3): l4759, July.-Sept.2017.
Artículo en Inglés | VETINDEX | ID: vti-16121

RESUMEN

In vitro maturation (IVM) of oocytes is a promising technology for both the treatment of human infertility and in animal production as a means of improving genetic gain. However, IVM derived oocytes remain inferior to those matured in vivo with reduced developmental potential. The environment in which an oocyte matures in vitro is vastly different to in vivo where maturation takes place within the ovarian follicle. The in vitro environment differs in oxygen concentration, exposure to light, and metabolite composition of culture media vs. follicle fluid, to name a few. Human follicle fluid contains the metabolite L-carnitine and has shown to be associated with human fertility. L-carnitine has known biological functions as an essential co-factor for beta-oxidation, regulating ATP production from lipids, and as a potent antioxidant. Importantly, it appears that cumulus cells and the oocyte lack the machinery to synthesize L-carnitinede novo. The inability for local production of L-carnitine during IVM and its importance in human fertility warrants investigation of its affects during IVM. The potential to improve oocyte quality by inclusion of L-carnitine in the culture media thus increasing the capacity for beta-oxidation and/or antioxidant activity of the culture media is receiving increased attention. This review Summarizes studies to date investigating the developmental importance of L-carnitine during IVM and the mechanisms by which improved developmental potential is elicited. Overall, the inclusion of L-carnitine during IVM of several species results in improved oocyte quality with increased development to blastocyst. This is likely due to the antioxidant capacity of L-carnitine and its ability to increase ATP production from intracellular lipid stores.(AU)


Asunto(s)
Animales , Carnitina/análisis , Carnitina/clasificación , Técnicas de Maduración In Vitro de los Oocitos/tendencias , Técnicas de Maduración In Vitro de los Oocitos/veterinaria , Antioxidantes
10.
Anim. Reprod. (Online) ; 14(3): l4759-475, July.-Sept.2017.
Artículo en Inglés | VETINDEX | ID: biblio-1461274

RESUMEN

In vitro maturation (IVM) of oocytes is a promising technology for both the treatment of human infertility and in animal production as a means of improving genetic gain. However, IVM derived oocytes remain inferior to those matured in vivo with reduced developmental potential. The environment in which an oocyte matures in vitro is vastly different to in vivo where maturation takes place within the ovarian follicle. The in vitro environment differs in oxygen concentration, exposure to light, and metabolite composition of culture media vs. follicle fluid, to name a few. Human follicle fluid contains the metabolite L-carnitine and has shown to be associated with human fertility. L-carnitine has known biological functions as an essential co-factor for beta-oxidation, regulating ATP production from lipids, and as a potent antioxidant. Importantly, it appears that cumulus cells and the oocyte lack the machinery to synthesize L-carnitinede novo. The inability for local production of L-carnitine during IVM and its importance in human fertility warrants investigation of its affects during IVM. The potential to improve oocyte quality by inclusion of L-carnitine in the culture media thus increasing the capacity for beta-oxidation and/or antioxidant activity of the culture media is receiving increased attention. This review Summarizes studies to date investigating the developmental importance of L-carnitine during IVM and the mechanisms by which improved developmental potential is elicited. Overall, the inclusion of L-carnitine during IVM of several species results in improved oocyte quality with increased development to blastocyst. This is likely due to the antioxidant capacity of L-carnitine and its ability to increase ATP production from intracellular lipid stores.


Asunto(s)
Animales , Carnitina/análisis , Carnitina/clasificación , Técnicas de Maduración In Vitro de los Oocitos/tendencias , Técnicas de Maduración In Vitro de los Oocitos/veterinaria , Antioxidantes
11.
Front Microbiol ; 8: 1428, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28824562

RESUMEN

Oleaginous microorganisms represent possible platforms for the sustainable production of oleochemicals and biofuels due to their metabolic robustness and the possibility to be engineered. Streptomyces coelicolor is among the narrow group of prokaryotes capable of accumulating triacylglycerol (TAG) as carbon and energy reserve. Although the pathways for TAG biosynthesis in this organism have been widely addressed, the set of genes required for their breakdown have remained elusive so far. Here, we identified and characterized three gene clusters involved in the ß-oxidation of fatty acids (FA). The role of each of the three different S. coelicolor FadAB proteins in FA catabolism was confirmed by complementation of an Escherichia coliΔfadBA mutant strain deficient in ß-oxidation. In S. coelicolor, the expression profile of the three gene clusters showed variation related with the stage of growth and the presence of FA in media. Flux balance analyses using a corrected version of the current S. coelicolor metabolic model containing detailed TAG biosynthesis reactions suggested the relevance of the identified fadAB genes in the accumulation of TAG. Thus, through the construction and analysis of fadAB knockout mutant strains, we obtained an S. coelicolor mutant that showed a 4.3-fold increase in the TAG content compared to the wild type strain grown under the same culture conditions.

12.
Curr Atheroscler Rep ; 19(9): 36, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28735349

RESUMEN

PURPOSE OF REVIEW: The quest for factors and mechanisms responsible for aberrant DNA methylation in human disease-including atherosclerosis-is a promising area of research. This review focuses on the role of fatty acids (FAs) as modulators of DNA methylation-in particular the role of mitochondrial beta-oxidation in FA-induced changes in DNA methylation during the progression of atherosclerosis. RECENT FINDINGS: Recent publications have advanced the knowledge in all areas touched by this review: the causal role of lipids in shaping the DNA methylome, the associations between chronic degenerative disease and mitochondrial function, the lipid composition of the atheroma, and the relevance of DNA hypermethylation in atherosclerosis. Evidence is beginning to emerge, linking the dynamics of FA type abundance, mitochondrial function, and DNA methylation in the atheroma and systemically. In particular, this review highlights mitochondrial beta-oxidation as an important regulator of DNA methylation in metabolic disease. Despite the many questions still unanswered, this area of research promises to identify mechanisms and molecular factors that establish a pathological gene expression pattern in atherosclerosis.


Asunto(s)
Aterosclerosis/etiología , Metilación de ADN , Ácidos Grasos/metabolismo , Mitocondrias/fisiología , Aterosclerosis/metabolismo , Humanos , Oxidación-Reducción
13.
Biochim Open ; 4: 19-30, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29450137

RESUMEN

Studies show that the continuous consumption of fructose can lead to nonalcoholic fatty liver disease (NAFLD) and steatohepatitis. We aimed to investigate the role of Metformin in an animal model of liver injury caused by fructose intake, focusing on the molecular markers of lipogenesis, beta-oxidation, and antioxidant defenses. Male three months old C57BL/6 mice were divided into control group (C) and fructose group (F, 47% fructose), maintained for ten weeks. After, the groups received Metformin or vehicle for a further eight weeks: control (C), control + Metformin (CM), fructose (F), and fructose + Metformin (FM). Fructose resulted in hepatic steatosis, insulin resistance and lower insulin sensitivity in association with higher mRNA levels of proteins linked with de novo lipogenesis and increased lipid peroxidation. Fructose diminished mRNA expression of antioxidant enzymes, and of proteins responsible for mitochondrial biogenesis. Metformin reduced de novo lipogenesis and increased the expression of proteins related to mitochondrial biogenesis, thereby increasing beta-oxidation and decreasing lipid peroxidation. Also, Metformin upregulated the expression and activity of antioxidant enzymes, providing a defense against increased reactive oxygen species generation. Therefore, a significant reduction in triglyceride accumulation in the liver, steatosis and lipid peroxidation was observed in the FM group. In conclusion, fructose increases de novo lipogenesis, reduces the antioxidant defenses, and diminishes mitochondrial biogenesis. After an extended period of fructose intake, Metformin treatment, even in continuing the fructose intake, can reverse, at least partially, the liver injury and prevents NAFLD progression to more severe states.

14.
Hepatol Res ; 47(9): 928-940, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27653239

RESUMEN

AIM: The aim of this study was to investigate the effects of rosuvastatin in a model of diet-induced obesity and non-alcoholic fatty liver disease, with attention to the activation of hepatic stellate cells (HSCs). METHOD: Male C57BL/6 mice received a control diet (C; 10% energy as lipids) or a high-fat diet (HF; 50% energy as lipids) for 12 weeks, followed by 7 weeks of treatment. Group CR received control diet + rosuvastatin; group HFR received high-fat diet + rosuvastatin. RESULTS: The HF group showed higher insulin, total cholesterol, triacylglycerol, and leptin levels than the C group, all of which were significantly diminished by rosuvastatin in the HFR group. The HF group had greater steatosis and activated HSCs than the C group, whereas rosuvastatin diminished the steatosis (less 21%, P < 0.001) and significantly inhibited the activation of the HSCs in the HFR group compared to the HF group. The sterol regulatory element-binding protein-1 and the peroxisome proliferator-activated receptor (PPAR)-γ protein expressions were increased in HF animals and reduced after treatment in the HFR group. By contrast, low PPAR-α and carnitine palmitoyltransferase-1 expressions were found in the HF group, and were restored by rosuvastatin treatment in the HFR group. CONCLUSION: Rosuvastatin mitigated hepatic steatosis by modulating PPAR balance, favoring PPAR-α over PPAR-γ downstream effects. The effects were accompanied by a diminishing of insulin resistance, the anti-inflammatory adipokine profile, and HSC activation, avoiding non-alcoholic fatty liver disease progression and non-alcoholic steatohepatitis onset in this model.

15.
Int. j. morphol ; 33(4): 1510-1517, Dec. 2015. ilus
Artículo en Inglés | LILACS | ID: lil-772346

RESUMEN

The high fat (HF) fed mothers may program susceptibility in offspring to chronic diseases and affect subsequent generations. The present study evaluated the liver structure in adulthood, focusing on the F1 and F2 generations. Females C57BL/6 (F0) were fed standard chow (SC) or HF diet (8 weeks) prior to mating and during the gestation and lactation to provide the F1 generation (SC-F1 and HF-F1). All other mothers and offspring fed SC. At 3 months old, F1 females were mated to produce the F2 generation (SC-F2 and HF-F2). The liver was kept in several fragments and prepared for histological analysis or frozen for biochemical and molecular analyzes. The F1 and F2 offspring were studied at 3 months old. HF-F1 had higher body mass (BM) compared to SC-F1 (P= 0.001), but not HF-F2 compared to SC-F2. HF-F1 had glucose intolerance when compared to SC-F1, but not HF-F2 compared to SC-F2. HF-F1 (P= 0.009) and HF-F2 (P= 0.03) showed hyperinsulinemia compared to their counterparts. Both groups HF-F1 and HF-F2 showed more steatosis than the SC counterparts (F1 and F2, P<0.0001). HF-F1 showed increased expression of PPAR-gamma and SREBP1-c compared to SC-F1 (P= 0.01). HF-F2 showed increased PPAR-gamma expression compared to SC-F2 (P= 0.04). In conclusion, HF-fed mother impairs both lipogenesis and beta-oxidation pathways in F1 through upregulation of PPAR-gamma and downregulation of PPAR-alpha. In F2, the only lipogenesis is enhanced, but it causes a disrupted PPAR balance, favoring the hepatic lipid accumulation and impaired metabolism in these animals that were not directly exposed to the maternal HF intake.


Los madres alimentadas con dieta rica en grasas (HF) pueden programar una susceptibilidad al desarrollo de enfermedades crónicas en su descendencia y de este modo afectar a las generaciones posteriores. El presente estudio evaluó la estructura del hígado en la edad adulta, centrándose en las generaciones F1 y F2. Las hembras C57BL/6 (F0) fueron alimentadas con dieta estándar (CS) o dieta HF (8 semanas) antes del apareamiento y durante la gestación y lactancia para producir la generación F1 (CS-F1 y HF-F1). Todas las demás madres y crías fueron alimentadas con CS. A los 3 meses de edad, las hembras F1 fueron apareadas para producir la generación F2 (CS-F2 y HF-F2). El hígado se conservó en varios fragmentos y se preparó, por un lado, para el análisis histológico, y por otro, se lo congeló para realizar análisis bioquímicos y moleculares. La descendencia F1 y F2 se estudió a los 3 meses de edad. HF-F1 tuvo una mayor masa corporal (BM) en comparación con CS-F1 (P= 0,001), pero no el grupo HF-F2 en comparación con CS-F2. HF-F1 tenía intolerancia a la glucosa en comparación con CS-F1, pero no el grupo HF-F2 en comparación con CS-F2. HF-F1 (P= 0,009) y HF-F2 (P= 0,03) mostraron hiperinsulinemia en comparación con sus homólogos. Ambos grupos HF-F1 y HF-F2 mostraron más esteatosis que las contrapartes CS (F1 y F2, P <0,0001). HF-F1 mostró una mayor expresión de PPAR-gamma y SREBP1-c en comparación con el grupo CS-F1 (P= 0,01). HF-F2 mostró aumento de la expresión de PPAR-gamma en comparación con CS-F2 (P= 0,04). En conclusión, la madre alimentada con HF presenta ambas vías afectadas, de lipogénesis y de la beta-oxidación, en la F1 a través de la regulación positiva de PPAR-gamma y con regulación a la baja de los PPAR-alfa. En F2, solo ha mejorado la vía de lipogénesis, pero causa un desbalance de PPAR, lo que favorece la acumulación de lípidos hepáticos y la alteración del metabolismo en estos animales que no estaban directamente expuestos a la ingesta materna de HF.


Asunto(s)
Animales , Masculino , Femenino , Embarazo , Ratones , Dieta Alta en Grasa/efectos adversos , Hígado Graso/patología , Obesidad/complicaciones , Animales Recién Nacidos , Western Blotting , Hiperinsulinismo , Lipogénesis , Ratones Endogámicos C57BL , Efectos Tardíos de la Exposición Prenatal
16.
World J Hepatol ; 7(8): 1012-9, 2015 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-26052390

RESUMEN

Lately, the world has faced tremendous progress in the understanding of non-alcoholic fatty liver disease (NAFLD) pathogenesis due to rising obesity rates. Peroxisome proliferator-activated receptors (PPARs) are transcription factors that modulate the expression of genes involved in lipid metabolism, energy homeostasis and inflammation, being altered in diet-induced obesity. Experimental evidences show that PPAR-alpha is the master regulator of hepatic beta-oxidation (mitochondrial and peroxisomal) and microsomal omega-oxidation, being markedly decreased by high-fat (HF) intake. PPAR-beta/delta is crucial to the regulation of forkhead box-containing protein O subfamily-1 expression and, hence, the modulation of enzymes that trigger hepatic gluconeogenesis. In addition, PPAR-beta/delta can activate hepatic stellate cells aiming to the hepatic recovery from chronic insult. On the contrary, PPAR-gamma upregulation by HF diets maximizes NAFLD through the induction of lipogenic factors, which are implicated in the fatty acid synthesis. Excessive dietary sugars also upregulate PPAR-gamma, triggering de novo lipogenesis and the consequent lipid droplets deposition within hepatocytes. Targeting PPARs to treat NAFLD seems a fruitful approach as PPAR-alpha agonist elicits expressive decrease in hepatic steatosis by increasing mitochondrial beta-oxidation, besides reduced lipogenesis. PPAR-beta/delta ameliorates hepatic insulin resistance by decreasing hepatic gluconeogenesis at postprandial stage. Total PPAR-gamma activation can exert noxious effects by stimulating hepatic lipogenesis. However, partial PPAR-gamma activation leads to benefits, mainly mediated by increased adiponectin expression and decreased insulin resistance. Further studies are necessary aiming at translational approaches useful to treat NAFLD in humans worldwide by targeting PPARs.

17.
Mol Nutr Food Res ; 59(8): 1573-83, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25913736

RESUMEN

SCOPE: Omega-3 polyunsaturated fatty acids (n-3 PUFA) found in fish oil activate PPAR-α, stimulate peroxisomal fatty acid (FA) ß-oxidation and prevent impairments on glucose homeostasis. METHODS AND RESULTS: Glucose metabolism and FA oxidation were studied in C57/Bl6 mice fed with diets containing either 3.6 and 31.5% fish oil or lard. To assess the effects of peroxisomal proliferation on FA oxidation independent of n-3 PUFA intake, mice were treated with the PPAR-α agonist WY-14643. n-3 PUFA-fed mice were protected from glucose intolerance and dyslipidemia compared to animals fed a lard-based high-fat diet. Most importantly, mice fed on the hyperlipidic diet based on fish oil as well as the WY-14643 treated mice showed twofold increase of odd, medium-chain, dicarboxylic acylcarnitines in the liver suggesting that not only ß-oxidation, but also α- and ω-oxidation of FA were increased. Finally, an oxidation assay using liver homogenates and palmitic acid as substrate revealed an over tenfold increased production of similar acylcarnitines, indicating that FA are their precursors. CONCLUSION: This study shows at the metabolite level that peroxisome proliferation induced either by fish oil or WY-14643 is associated with increased α- and ω-oxidation of FA producing specific acylcarnitines that can be utilized as biomarkers of peroxisomal FA oxidation.


Asunto(s)
Carnitina/análogos & derivados , Dieta Alta en Grasa/efectos adversos , Grasas Insaturadas en la Dieta/metabolismo , Ácidos Grasos Omega-3/metabolismo , Hígado/metabolismo , Sobrepeso/metabolismo , Peroxisomas/metabolismo , Animales , Biomarcadores/química , Biomarcadores/metabolismo , Carnitina/química , Carnitina/metabolismo , Grasas de la Dieta/efectos adversos , Grasas Insaturadas en la Dieta/efectos adversos , Grasas Insaturadas en la Dieta/uso terapéutico , Ácidos Grasos Omega-3/uso terapéutico , Aceites de Pescado/uso terapéutico , Regulación de la Expresión Génica/efectos de los fármacos , Intolerancia a la Glucosa/etiología , Intolerancia a la Glucosa/prevención & control , Hiperlipidemias/etiología , Hiperlipidemias/prevención & control , Hígado/efectos de los fármacos , Hígado/enzimología , Masculino , Ratones Endogámicos C57BL , Peso Molecular , Sobrepeso/etiología , Sobrepeso/fisiopatología , Sobrepeso/prevención & control , Oxidación-Reducción , Proliferadores de Peroxisomas/farmacología , Peroxisomas/efectos de los fármacos , Peroxisomas/enzimología , Pirimidinas/farmacología
18.
Iatreia ; Iatreia;27(4): 410-416, oct.-dic. 2014. ilus, tab
Artículo en Español | LILACS | ID: lil-726838

RESUMEN

Introducción: la deficiencia de acil-CoA deshidrogenasa de cadena muy larga (VLCAD) se manifiesta en los primeros meses de vida con cardiomiopatía hipertrófica o dilatada y arritmias, que pueden llegar a ser letales. Objetivo: analizar la producción de metabolitos en fibroblastos de pacientes con deficiencia de VLCAD y cardiomiopatía, incubados con ácido oleico deuterado. Materiales y métodos: se incubaron fibroblastos de cuatro pacientes con deficiencia de VLCAD y cardiomiopatía y 10 de controles en presencia de ácido oleico deuterado. Resultados y conclusión: en pacientes con esta deficiencia se encontró un perfil característico luego de la incubación de los fibroblastos. Esta prueba se podría usar para confirmar in vitro la deficiencia de VLCAD con cardiomiopatía.


Introduction: Deficiency of VLCAD is present in the early months of life, with hypertrophic or dilated cardiomyopathy and arrhythmias which may be lethal. Objective: To analize the production of metabolites in fibroblasts from patientes with VLCAD deficiency and cardiomyopathy incubated with deuterated oleic acid. Materials and methods: Fibroblasts from four patients with VLCAD deficiency and cardiomyopathy and from ten controls were incubated in the presence of deuterated oleic acid. Results and conclusison: A characteristic profile was found in the fibroblastos of patients with VLCAD after the described incubation. This test could be used to confirm VLCAD deficiency with cardiomyopathy.


Introdução: a deficiência de acil-CoA desidrogenase de corrente muito longa (VLCAD) manifesta-se nos primeiros meses de vida com cardiomiopatia hipertrófica ou dilatada e arritmias, que podem chegar a ser letais. Objetivo: analisar a produção de metabólitos em fibroblastos de pacientes com deficiência de VLCAD e cardiomiopatia, incubados com ácido oleico deuterado. Materiais e métodos: incubaram-se fibroblastos de quatro pacientes com deficiência de VLCAD e cardiomiopatia e 10 de controles em presença de acido oleico deuterado. Resultados e conclusão: em pacientes com esta deficiência se encontrou um perfil característico depois da encubação dos fibroblastos. Esta prova se poderia usar para confirmar in vitro a deficiência de VLCAD com cardiomiopatia.


Asunto(s)
Humanos , Acil-CoA Deshidrogenasa de Cadena Larga/deficiencia , Cardiomiopatías , Fibroblastos/metabolismo
19.
Acta méd. costarric ; 56(3): 134-137, jul.-sep. 2014. ilus
Artículo en Español | LILACS | ID: lil-715380

RESUMEN

Paciente pediátrico, con el diagnóstico de distrofia muscular de Duchenne y deficiencia de acil-CoA deshidrogenasa de cadenas medias, ambas patologías confirmadas por medio de análisis molecular, al detectarse la deleción de los exones 45 al 50 en el gen DMD, y la mutación A985G en estado homocigoto del gen ACADM...


Asunto(s)
Humanos , Masculino , Recién Nacido , Lactante , Preescolar , Ácidos Grasos , Distrofia Muscular de Duchenne
20.
Iatreia ; Iatreia;24(3): 267-271, sept.-nov. 2011. tab
Artículo en Español | LILACS | ID: lil-600391

RESUMEN

Introduccion: la aciduria glutarica tipo II, o deficiencia multiple de acil-CoA deshidrogenasas,es un trastorno causado por deficiencia de la flavoproteina de transferencia de electrones,de su oxidorreductasa o de ambas; se trata de una enfermedad metabolica autosomica recesiva, caracterizada por acidosis, hipoglicemia, aciduria organica, olor a pies sudados y malformaciones en cerebro y riñones. Objetivo: analizar las tasas de oxidacion de sustratos tritiados por fibroblastos de pacientescon aciduria glutarica tipo II. Materiales y metodos: se incubaron fibroblastos de dos pacientes con aciduria glutarica tipoII y de 20 controles en presencia de acidos palmitico y miristico tritiados. Resultados: se encontro muy deprimida (16%-18%) la oxidacion de los sustratos tritiados porlos fibroblastos procedentes de pacientes con aciduria glutarica tipo II en comparacion con los controles. Conclusion: la prueba estudiada permite la confirmacion in vitro del diagnostico de aciduriaglutarica tipo II.


Introduction: Glutaric aciduria type II (GA II), or multiple acyl-CoA dehydrogenase deficiency, is a disorder caused by deficiency of either electron transport flavoprotein or electron transport flavoprotein oxyreductase. It is an autsomal recessive metabolic disease, characterized by acidosis, hypoglycemia, organic aciduria, sweat-sock odour, and malformations in brain and kidneys. Objective: To analyse the oxidation rate of tritiated substrates by fibroblasts of patients with GA II. Materials and methods: Fibroblasts of two patients with GA II were incubated with tritiated palmitic and myristic acids. Results: Oxidation of tritiated substrates by fibroblasts of patients with GA II was very depressed (16%-18%) in comparison with controls. Conclusion: Diagnosis of GA II may be confirmed in vitro by the studied test.


Asunto(s)
Humanos , Acetilcoenzima A , Ácidos Grasos , Oxidorreductasas/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA