Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Environ Manage ; 344: 118475, 2023 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-37406491

RESUMEN

Second generation biofuel crop Miscanthus x giganteus (Mxg) was studied as a candidate for petroleum hydrocarbons (PHs) contaminated soil phytomanagement. The soil was polluted by diesel in wide concentration gradient up to 50 g⋅kg-1 in an ex-situ pot experiment. The contaminated soil/plant interactions were investigated using plant biometric and physiological parameters, soil physico-chemical and microbial community's characteristics. The plant parameters and chlorophyll fluorescence indicators showed an inhibitory effect of diesel contamination; however much lower than expected from previously published results. Moreover, lower PHs concentrations (5 and 10 g⋅kg-1) resulted in positive reinforcement of electron transport as a result of hormesis effect. The soil pH did not change significantly during the vegetation season. The decrease of total organic carbon was significantly lower in planted pots. Soil respiration and dehydrogenases activity increased with the increasing contamination indicating ongoing PHs biodegradation. In addition, microbial biomass estimated by phospholipid fatty acids increased only at higher PHs concentrations. Higher dehydrogenases values were obtained in planted pots compared to unplanted. PHs degradation followed the first-order kinetics and for the middle range of contamination (10-40 g⋅kg-1) significantly lower PHs half-lives were determined in planted than unplanted soil pointing on phytoremediation. Diesel degradation was in range 35-70 % according to pot variant. Results confirmed the potential of Mxg for diesel contaminated soils phytomanagement mainly in PHs concentrations up to 20 g⋅kg-1 where phytoremediation was proved, and biomass yield was reduced only by 29 %.


Asunto(s)
Petróleo , Contaminantes del Suelo , Biodegradación Ambiental , Contaminantes del Suelo/análisis , Poaceae/metabolismo , Plantas/metabolismo , Hidrocarburos/metabolismo , Suelo , Oxidorreductasas/metabolismo
2.
J Diabetes Res ; 2022: 4587907, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36147256

RESUMEN

Introduction: Monounsaturated fatty acids (MUFA) are understood to have therapeutic and preventive effects on chronic complications associated with type 2 diabetes mellitus (T2DM); however, there are differences between individual MUFAs. Although the effects of palmitoleic acid (POA) are still debated, POA can regulate glucose homeostasis, lipid metabolism, and cytokine production, thus improving metabolic disorders. In this study, we investigated and compared the metabolic effects of POA and oleic acid (OA) supplementation on glucose and lipid metabolism, insulin sensitivity, and inflammation in a prediabetic model, the hereditary hypertriglyceridemic rat (HHTg). HHTg rats exhibiting genetically determined hypertriglyceridemia, insulin resistance, and impaired glucose tolerance were fed a standard diet. POA and OA were each administered intragastrically at a dose of 100 mg/kg b.wt. for four weeks. Results: Supplementation with both MUFAs significantly elevated insulin and glucagon levels, but only POA decreased nonfasting glucose. POA-treated rats showed elevated circulating NEFA associated with increased lipolysis, lipoprotein lipase gene expression, and fatty acid reesterification in visceral adipose tissue (VAT). The mechanism of improved insulin sensitivity of peripheral tissues (measured as insulin-stimulated lipogenesis and glycogenesis) in POA-treated HHTg rats could contribute increased circulating adiponectin and omentin levels together with elevated FADS1 gene expression in VAT. POA-supplemented rats exhibited markedly decreased proinflammatory cytokine production by VAT, which can alleviate chronic inflammation. OA-supplemented rats exhibited decreased arachidonic acid (AA) profiles and decreased proinflammatory AA-derived metabolites (20-HETE) in membrane phospholipids of peripheral tissues. Slightly increased FADS1 gene expression after OA along with increased adiponectin production by VAT was reflected in slightly ameliorated adipose tissue insulin sensitivity (increased insulin-stimulated lipogenesis). Conclusions: Our results show that POA served as a lipokine, ameliorating insulin sensitivity in peripheral tissue and markedly modulating the metabolic activity of VAT including cytokine secretion. OA had a beneficial effect on lipid metabolism and improved inflammation by modulating AA metabolism.


Asunto(s)
Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Estado Prediabético , Adiponectina , Animales , Antiinflamatorios , Ácidos Araquidónicos , Citocinas , Ácidos Grasos/metabolismo , Ácidos Grasos Monoinsaturados/farmacología , Ácidos Grasos Monoinsaturados/uso terapéutico , Ácidos Grasos no Esterificados , Glucagón , Glucosa/metabolismo , Inflamación , Insulina/metabolismo , Lipoproteína Lipasa , Ácido Oléico/farmacología , Estado Prediabético/tratamiento farmacológico , Ratas
3.
Antioxidants (Basel) ; 10(3)2021 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-33809515

RESUMEN

The selenium (Se) enrichment of yeasts and lactic acid bacteria (LAB) has recently emerged as a novel concept; the individual health effects of these beneficial microorganisms are combined by supplying the essential micronutrient Se in a more bioavailable and less toxic form. This study investigated the bioavailability of Se in the strains Enterococcus faecium CCDM 922A (EF) and Streptococcus thermophilus CCDM 144 (ST) and their respective Se-enriched forms, SeEF and SeST, in a CD (SD-Sprague Dawley) IGS rat model. Se-enriched LAB administration resulted in higher Se concentrations in the liver and kidneys of rats, where selenocystine was the prevalent Se species. The administration of both Se-enriched strains improved the antioxidant status of the animals. The effect of the diet was more pronounced in the heart tissue, where a lower glutathione reductase content was observed, irrespective of the Se fortification in LAB. Interestingly, rats fed diets with EF and SeEF had higher glutathione reductase activity. Reduced concentrations of serum malondialdehyde were noted following Se supplementation. Diets containing Se-enriched strains showed no macroscopic effects on the liver, kidneys, heart, and brain and had no apparent influence on the basic parameters of the lipid metabolism. Both the strains tested herein showed potential for further applications as promising sources of organically bound Se and Se nanoparticles.

4.
Pharmaceutics ; 14(1)2021 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-35056941

RESUMEN

The combination of plant-derived compounds with anti-diabetic agents to manage hepatic steatosis closely associated with diabetes mellitus may be a new therapeutic approach. Silymarin, a complex of bioactive substances extracted from Silybum marianum, evinces an antioxidative, anti-inflammatory, and hepatoprotective activity. In this study, we investigated whether metformin (300 mg/kg/day for four weeks) supplemented with micronized silymarin (600 mg/kg/day) would be effective in mitigating fatty liver disturbances in a pre-diabetic model with dyslipidemia. Compared with metformin monotherapy, the metformin-silymarin combination reduced the content of neutral lipids (TAGs) and lipotoxic intermediates (DAGs). Hepatic gene expression of enzymes and transcription factors involved in lipogenesis (Scd-1, Srebp1, Pparγ, and Nr1h) and fatty acid oxidation (Pparα) were positively affected, with hepatic lipid accumulation reducing as a result. Combination therapy also positively influenced arachidonic acid metabolism, including its metabolites (14,15-EET and 20-HETE), mitigating inflammation and oxidative stress. Changes in the gene expression of cytochrome P450 enzymes, particularly Cyp4A, can improve hepatic lipid metabolism and moderate inflammation. All these effects play a significant role in ameliorating insulin resistance, a principal background of liver steatosis closely linked to T2DM. The additive effect of silymarin in metformin therapy can mitigate fatty liver development in the pre-diabetic state and before the onset of diabetes.

5.
Clin Nutr ; 40(4): 1822-1833, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33081982

RESUMEN

BACKGROUND & AIMS: Reward circuitry in the brain plays a key role in weight regulation. We tested the effects of a plant-based meal on these brain regions. METHODS: A randomized crossover design was used to test the effects of two energy- and macronutrient-matched meals: a vegan (V-meal) and a conventional meat (M-meal) on brain activity, gastrointestinal hormones, and satiety in participants with type 2 diabetes (T2D; n = 20), overweight/obese participants (O; n = 20), and healthy controls (H; n = 20). Brain perfusion was measured, using arterial spin labeling functional brain imaging; satiety was assessed using a visual analogue scale; and plasma concentrations of gut hormones were determined at 0 and 180 min. Repeated-measures ANOVA was used for statistical analysis. Bonferroni correction for multiple comparisons was applied. The Hedge's g statistic was used to measure the effect size for means of paired difference between the times (180-0 min) and meal types (M-V meal) for each group. RESULTS: Thalamus perfusion was the highest in patients with T2D and the lowest in overweight/obese individuals (p = 0.001). Thalamus perfusion decreased significantly after ingestion of the M-meal in men with T2D (p = 0.04) and overweight/obese men (p = 0.004), and it decreased significantly after ingestion of the V-meal in healthy controls (p < 0.001; Group x Meal x Time: F = 3.4; p = 0.035). The effect size was -0.41 (95% CI, -1.14 to 0.31; p = 0.26) for men with diabetes; -0.72 (95% CI, -1.48 to 0.01; p = 0.05) for overweight/obese men; and 0.82 (95% CI, 0.09 to 1.59; p = 0.03) for healthy men. Postprandial secretion of active GLP-1 increased after the V-meal compared with the M-meal by 42% (95% CI 25-62%; p = 0.003) in men with T2D and by 41% (95% CI 24-61%; p = 0.002) in healthy controls. Changes in thalamus perfusion after ingestion of both test meals correlated with changes in satiety (r = +0.68; p < 0.01), fasting plasma insulin (r = +0.40; p < 0.01), C-peptide (r = +0.48; p < 0.01) and amylin (r = +0.55; p < 0.01), and insulin secretion at 5 mmol/l (r = +0.77; p < 0.05). CONCLUSIONS: The higher postprandial GLP-1 secretion after the V-meal in men with T2D, with concomitant greater satiety and changes in thalamus perfusion, suggest a potential use of plant-based meals in addressing the key pathophysiologic mechanisms of food intake regulation. Trial registration ClinicalTrials.gov number, NCT02474147.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Dieta Vegetariana/métodos , Ingestión de Energía , Nutrientes/metabolismo , Sobrepeso/metabolismo , Tálamo/irrigación sanguínea , Adulto , Anciano , Estudios Cruzados , Dieta/métodos , Humanos , Masculino , Comidas , Persona de Mediana Edad , Obesidad/metabolismo , Tálamo/metabolismo
6.
Nutrients ; 12(8)2020 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-32781598

RESUMEN

Omega-3 polyunsaturated fatty acids (ω-3PUFAs) are introduced into parenteral nutrition (PN) as hepatoprotective but may be susceptible to the lipid peroxidation while olive oil (OO) is declared more peroxidation resistant. We aimed to estimate how the lipid composition of PN mixture affects plasma and erythrocyte lipidome and the propensity of oxidative stress. A cross-sectional comparative study was performed in a cohort of adult patients who were long-term parenterally administered ω-3 PUFAs without (FO/-, n = 9) or with (FO/OO, n = 13) olive oil and healthy age- and sex-matched controls, (n = 30). Lipoperoxidation assessed as plasma and erythrocyte malondialdehyde content was increased in both FO/- and FO/OO groups but protein oxidative stress (protein carbonyls in plasma) and low redox status (GSH/GSSG in erythrocytes) was detected only in the FO/- subcohort. The lipidome of all subjects receiving ω-3 PUFAs was enriched with lipid species containing ω-3 PUFAs (FO/-˃FO/OO). Common characteristic of all PN-dependent patients was high content of fatty acyl-esters of hydroxy-fatty acids (FAHFAs) in plasma while acylcarnitines and ceramides were enriched in erythrocytes. Plasma and erythrocyte concentrations of plasmanyls and plasmalogens (endogenous antioxidants) were decreased in both patient groups with a significantly more pronounced effect in FO/-. We confirmed the protective effect of OO in PN mixtures containing ω-3 PUFAs.


Asunto(s)
Antioxidantes/metabolismo , Emulsiones Grasas Intravenosas/farmacología , Ácidos Grasos Omega-3/farmacología , Estrés Oxidativo/efectos de los fármacos , Nutrición Parenteral/métodos , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Estudios Transversales , Eritrocitos/metabolismo , Femenino , Aceites de Pescado/farmacología , Humanos , Enfermedades Intestinales/sangre , Enfermedades Intestinales/terapia , Lipidómica , Lípidos/sangre , Masculino , Persona de Mediana Edad , Aceite de Oliva/farmacología , Nutrición Parenteral/efectos adversos
7.
Biomed Res Int ; 2019: 7084734, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30941370

RESUMEN

Butyrate produced by the intestinal microbiota is essential for proper functioning of the intestinal immune system. Total dependence on parenteral nutrition (PN) is associated with numerous adverse effects, including severe microbial dysbiosis and loss of important butyrate producers. We hypothesised that a lack of butyrate produced by the gut microbiota may be compensated by its supplementation in PN mixtures. We tested whether i.v. butyrate administration would (a) positively modulate intestinal defence mechanisms and (b) counteract PN-induced dysbiosis. Male Wistar rats were randomised to chow, PN, and PN supplemented with 9 mM butyrate (PN+But) for 12 days. Antimicrobial peptides, mucins, tight junction proteins, and cytokine expression were assessed by RT-qPCR. T-cell subpopulations in mesenteric lymph nodes (MLN) were analysed by flow cytometry. Microbiota composition was assessed in caecum content. Butyrate supplementation resulted in increased expression of tight junction proteins (ZO-1, claudin-7, E-cadherin), antimicrobial peptides (Defa 8, Rd5, RegIIIγ), and lysozyme in the ileal mucosa. Butyrate partially alleviated PN-induced intestinal barrier impairment and normalised IL-4, IL-10, and IgA mRNA expression. PN administration was associated with an increase in Tregs in MLN, which was normalised by butyrate. Butyrate increased the total number of CD4+ and decreased a relative amount of CD8+ memory T cells in MLN. Lack of enteral nutrition and PN administration led to a shift in caecal microbiota composition. Butyrate did not reverse the altered expression of most taxa but did influence the abundance of some potentially beneficial/pathogenic genera, which might contribute to its overall beneficial effect.


Asunto(s)
Butiratos/farmacología , Suplementos Dietéticos , Microbioma Gastrointestinal , Intestinos/patología , Nutrición Parenteral , Animales , Biodiversidad , Colon/efectos de los fármacos , Colon/patología , Microbioma Gastrointestinal/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Íleon/efectos de los fármacos , Íleon/patología , Intestino Delgado/efectos de los fármacos , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/metabolismo , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Masculino , Modelos Animales , Mucinas/biosíntesis , Células de Paneth/efectos de los fármacos , Células de Paneth/metabolismo , Péptidos/genética , Péptidos/metabolismo , Permeabilidad , Fenotipo , Filogenia , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Wistar , Proteínas de Uniones Estrechas/metabolismo
8.
Xenobiotica ; 45(9): 751-6, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26068528

RESUMEN

1. To compare the effectiveness of different drug forms of silymarin: standardized extract of silymarin (SS), micronized silymarin (MS) and silymarin in the form of phytosome (PS) on dyslipidemia and liver fat accumulation in a model of metabolic syndrome, in non-obese hereditary hypertriglyceridemic rats. The second aim of this study was to slightly uncover the silymarin action on enzymes and proteins involved in cholesterol metabolism and excretion. 2. Silymarin administered to hereditary hypertriglyceridemic rats as dietary supplements (1%) for 4 weeks significantly lowered the plasma levels of triglycerides, total cholesterol and markedly increased HDL cholesterol level. Western blot analyses showed significant increase in the protein expression of CYP7A1 and CYP4A and increase in protein expression of selected ABC transporters. Silymarin in the form of phytosome and micronized silymarin were more effective forms of silymarin. 3. These findings suggest that silymarin may favorably affect the metabolism of cholesterol and triglycerides in rats with metabolic syndrome. Raising HDL levels suggests potentially important anti-atherogenic effect of silymarin. The changes in expression of cytochromes P450 and ABC transporters involved in cholesterol metabolism and excretion could be partially responsible for the hypolipidemic effect of silymarin.


Asunto(s)
Dislipidemias/complicaciones , Dislipidemias/tratamiento farmacológico , Síndrome Metabólico/complicaciones , Síndrome Metabólico/tratamiento farmacológico , Silimarina/uso terapéutico , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Disponibilidad Biológica , Western Blotting , Colesterol 7-alfa-Hidroxilasa/metabolismo , Citocromo P-450 CYP4A/metabolismo , Dislipidemias/sangre , Lípidos/sangre , Hígado/efectos de los fármacos , Hígado/enzimología , Síndrome Metabólico/sangre , Ratas Wistar , Silimarina/farmacología
9.
Nutrition ; 31(7-8): 1045-51, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26059381

RESUMEN

OBJECTIVE: The metabolic health effects of conjugated linoleic acid (CLA), which is one of the principal polyunsaturated fatty acids, are controversial and still not fully accepted. The aim of this study was to examine the effects of CLA on adiposity, ectopic lipid accumulation, and insulin-resistant states in a metabolic syndrome model of non-obese hereditary rats with hypertriacylglycerolmia (HHTg). METHODS: Groups of adult male HHTg rats were fed a high-carbohydrate diet (70% sucrose) with a 2% mixture of CLA isomers, or with the same amount of sunflower oil (control group) for 2 mo. RESULTS: CLA supplementation decreased body weight gain (P < 0.05) and visceral adipose tissue weight (P < 0.01), and distinctively reduced serum triacylglycerols (P < 0.01) and triacylglycerol accumulation in the liver, heart, muscle, and aorta. CLA-treated rats exhibited increased insulin sensitivity in the adipose (P < 0.01), a higher release of fatty acids (P < 0.001), and increased adiponectin secretion (P < 0.01).In the skeletal muscle, CLA supplementation was associated with increased glucose oxidation (P < 0.01) and an elevated anti-inflammatory index (P < 0.05), according to phospholipid fatty acid composition. In the liver, CLA reduced the oxidized form of glutathione and elevated the activity of glutathione-dependent antioxidant enzymes. CONCLUSION: Results suggest that CLA supplementation may protect against HHTg-induced dyslipidemia, ectopic lipid deposition, and insulin resistance. Increased glucose oxidation in the skeletal muscle as well as adiponectin secretion may play a role in the mechanism of the CLA action. Results suggest that CLA could reduce the negative consequences of HHTg and metabolic syndrome.


Asunto(s)
Suplementos Dietéticos , Hipertrigliceridemia/dietoterapia , Resistencia a la Insulina , Ácidos Linoleicos Conjugados/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Carbohidratos de la Dieta/administración & dosificación , Carbohidratos de la Dieta/efectos adversos , Modelos Animales de Enfermedad , Glucosa/metabolismo , Hipertrigliceridemia/complicaciones , Hipertrigliceridemia/etiología , Insulina/sangre , Ácidos Linoleicos Conjugados/administración & dosificación , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Oxidación-Reducción/efectos de los fármacos , Ratas
10.
Appl Physiol Nutr Metab ; 40(3): 280-91, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25723909

RESUMEN

The aim of this study was to estimate the effect of carnitine supplementation on lipid disorders and peripheral tissue insulin sensitivity in a non-obese animal model of insulin resistance, the hereditary hypertriglyceridemic (HHTg) rat. Male HHTg rats were fed a standard diet, and half of them received daily doses of carnitine (500 mg·kg(-1) body weight) for 8 weeks. Rats of the original Wistar strain were used for comparison. HHTg rats exhibited increased urinary excretion of free carnitine and reduced carnitine content in the liver and blood. Carnitine supplementation compensated for this shortage and promoted urinary excretion of acetylcarnitine without any signs of (acyl)carnitine accumulation in skeletal muscle. Compared with their untreated littermates, carnitine-treated HHTg rats exhibited lower weight gain, reduced liver steatosis, lower fasting triglyceridemia, and greater reduction of serum free fatty acid content after glucose load. Carnitine treatment was associated with increased mitochondrial biogenesis and oxidative capacity for fatty acids, amelioration of oxidative stress, and restored substrate switching in the liver. In skeletal muscle (diaphragm), carnitine supplementation was associated with significantly higher palmitate oxidation and a more favorable complete to incomplete oxidation products ratio. Carnitine supplementation further enhanced insulin sensitivity ex vivo. No effects on whole-body glucose tolerance were observed. Our data suggest that some metabolic syndrome-related disorders, particularly fatty acid oxidation, steatosis, and oxidative stress in the liver, could be attenuated by carnitine supplementation. The effect of carnitine could be explained, at least partly, by enhanced substrate oxidation and increased fatty acid transport from tissues in the form of short-chain acylcarnitines.


Asunto(s)
Carnitina/farmacología , Hipertrigliceridemia/genética , Metabolismo de los Lípidos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Animales , Carnitina/administración & dosificación , Carnitina/análogos & derivados , Carnitina/sangre , Carnitina/metabolismo , Carnitina/orina , ADN Mitocondrial/genética , Suplementos Dietéticos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Predisposición Genética a la Enfermedad , Homeostasis , Hipertrigliceridemia/metabolismo , Resistencia a la Insulina , Riñón/efectos de los fármacos , Riñón/metabolismo , Hígado/metabolismo , Masculino , Músculo Esquelético/metabolismo , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA