Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 103
Filtrar
2.
Handb Exp Pharmacol ; 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38421444

RESUMEN

Type 2 diabetes (T2D) is a disease that occurs when cells do not respond normally to insulin, a condition called insulin resistance, which leads to high blood glucose levels. Although it can be treated pharmacologically, dietary habits beyond carbohydrate restriction can be highly relevant in the management of T2D. Emerging evidence supports the possibility that natural products (NPs) could contribute to managing blood glucose or counteract the undesirable effects of hyperglycemia and insulin resistance. This chapter summarizes the relevant preclinical evidence involving the flavonoid (-)-epicatechin (EC) in the optimization of glucose homeostasis, reducing insulin resistance and/or diabetes-associated disorders. Major effects of EC are observed on (i) intestinal functions, including digestive enzymes, glucose transporters, microbiota, and intestinal permeability, and (ii) redox homeostasis, including oxidative stress and inflammation. There is still a need for further clinical studies to confirm the in vitro and rodent data, allowing recommendations for EC, particularly in prediabetic and T2D patients. The collection of similar data and the lack of clinical evidence for EC is also applicable to other NPs.

3.
Redox Biol ; 67: 102927, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37857000

RESUMEN

Nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOXs) are enzymes that generate superoxide anion (O2•-) and hydrogen peroxide (H2O2), and that are widely distributed in mammalian tissues. Many bioactives, especially plant (poly)phenols are being studied for their capacity to regulate NOXs. The modulation of these enzymes are of central relevance to maintain redox homeostasis and regulate cell signaling. In in vitro and ex vivo assays, and in experimental animal models, different (poly)phenols are able to modulate NOX-dependent generation of O2•- and H2O2. Mechanistically, most of the known effects of (poly)phenols and of their metabolites on NOX1, NOX2, and NOX4, include the modulation of: i) the expression of the different constituent subunits, and/or ii) posttranslational modifications involved in the assembly and translocation of the protein complexes. Very limited evidence is available on a direct action of (poly)phenols on NOX active site (electron-transferring protein). Moreover, it is suggested that the regulation by (poly)phenols of systemic events, e.g. inflammation, is frequently associated with their capacity to regulate NOX activation. Although of physiological significance, more studies are needed to understand the specific targets/mechanisms of NOX regulation by (poly)phenols, and the (poly)phenol chemical structures and moieties directly involved in the observed effects. It should be kept in mind the difficulties of NOX's studies associated with the complexity of NOXs biochemistry and the methodological limitations of O2•- and H2O2 the determinations. Studies relating human ingestion of specific (poly)phenols, with NOX activity and disease conditions, are guaranteed to better understand the health importance of (poly)phenol consumption and the involvement of NOXs as biological targets.


Asunto(s)
Peróxido de Hidrógeno , Fenoles , Animales , Humanos , Especies Reactivas de Oxígeno/metabolismo , Peróxido de Hidrógeno/metabolismo , Fenol , NADPH Oxidasas/metabolismo , NADPH Oxidasa 1 , Mamíferos/metabolismo
5.
Mol Aspects Med ; 89: 101168, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36610861
6.
Mol Aspects Med ; 89: 101158, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36517273

RESUMEN

Nitric oxide (•NO) is an essential molecule able to control and regulate many biological functions. Additionally, •NO bears a potential toxicity or damaging effects under conditions of uncontrolled production, and because of its participation in redox-sensitive pathways and oxidizing reactions. Several plant (poly)phenols present in the diet are able to regulate the enzymes producing •NO (NOSs). In addition, (poly)phenols are implicated in defining •NO bioavailability, especially by regulating NADPH oxidases (NOXs), and the subsequent generation of superoxide and •NO depletion. Nitrolipids are compounds that are present in animal tissues because of dietary consumption, e.g. of olive oil, and/or as result of endogenous production. This endogenous production of nitrolipids is dependent on the nitrate/nitrite presence in the diet. Select nitrolipids, e.g. the nitroalkenes, are able to exert •NO-like signaling actions, and act as •NO reservoirs, becoming relevant for systemic •NO bioavailability. Furthermore, the presence of (poly)phenols in the stomach reduces dietary nitrite to •NO favoring nitrolipids formation. In this review we focus on the capacity of molecules representing these two groups of bioactives, i.e. (poly)phenols and nitrolipids, as relevant participants in •NO metabolism and bioavailability. This participation acquires especial relevance when human homeostasis is lost, for example under inflammatory conditions, in which the protective actions of (poly)phenols and/or nitrolipids have been associated with local and systemic •NO bioavailability.


Asunto(s)
Nitritos , Fenoles , Animales , Humanos , Nitritos/metabolismo , Nitratos , Óxido Nítrico/metabolismo , Dieta
7.
Mol Aspects Med ; 89: 101156, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36379746

RESUMEN

Anthocyanins (AC) are flavonoids abundant in the human diet, which consumption has been associated to several health benefits, including the mitigation of cardiovascular disease, type 2 diabetes, non-alcoholic fatty liver disease, and neurological disorders. It is widely recognized that the gastrointestinal (GI) tract is not only central for food digestion but actively participates in the regulation of whole body physiology. Given that AC, and their metabolites reach high concentrations in the intestinal lumen after food consumption, their biological actions at the GI tract can in part explain their proposed local and systemic health benefits. In terms of mechanisms of action, AC have been found to: i) inhibit GI luminal enzymes that participate in the absorption of lipids and carbohydrates; ii) preserve intestinal barrier integrity and prevent endotoxemia, inflammation and oxidative stress; iii) sustain goblet cell number, immunological functions, and mucus production; iv) promote a healthy microbiota; v) be metabolized by the microbiota to AC metabolites which will be absorbed and have systemic effects; and vi) modulate the metabolism of GI-generated hormones. This review will summarize and discuss the latest information on AC actions at the GI tract and their relationship to overall health benefits.


Asunto(s)
Antocianinas , Diabetes Mellitus Tipo 2 , Humanos , Diabetes Mellitus Tipo 2/metabolismo , Tracto Gastrointestinal/metabolismo , Flavonoides/metabolismo , Dieta
8.
Free Radic Biol Med ; 188: 71-82, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35691508

RESUMEN

Consumption of high fat diets (HFD) mimics a modern or "Western style" diet pattern and can impair intestinal barrier integrity, leading to endotoxemia and associated unhealthy conditions. This study investigated if supplementation with an anthocyanin (cyanidin and delphinidin glucosides)-rich extract (CDRE) could revert or mitigate HFD-induced alterations of colonic physiology in part through the regulation of Toll-Like Receptor 4 (TLR-4)- and redox-regulated signaling. C57BL/6J male mice were fed for 4 weeks with a control or an HFD. Then, mice were divided in four groups fed either control or HFD, or these diets supplemented with CDRE for the subsequent 4 weeks. After 8 weeks on the HFD we observed in the colon: i) disruption of tight junction structure and function; ii) increased TLR-4 expression; iii) increased NADPH oxidase NOX1 expression, and iv) activation of redox-sensitive and TLR-4-triggered pathways, i.e. NF-κB, ERK1/2, JNK1/2, PI3K/Akt. All these events were prevented or reverted by CDRE supplementation. Supporting the relevance of CDRE-mediated downregulation of TLR-4 on its colon beneficial effect; in vitro (Caco-2 cell monolayers), cyanidin, delphinidin and their metabolites protocatechuic and gallic acid, mitigated lipopolysaccharide (LPS)-induced monolayer permeabilization by restoring tight junction structure and dynamics and preventing lipid/protein oxidation. The CDRE also mitigated HFD-mediated alterations in parameters of goblet cell differentiation and function, including the downregulation of markers of goblet cell differentiation (Klf4), and intestinal mucosa healing (Tff3). Results show that a short-term supplementation with cyanidin and delphinidin, protect from HFD-induced alterations in colon physiology in part through the modulation of TLR-4- and redox-regulated signaling.


Asunto(s)
Antocianinas , Dieta Alta en Grasa , Animales , Antocianinas/metabolismo , Antocianinas/farmacología , Células CACO-2 , Colon/metabolismo , Dieta Alta en Grasa/efectos adversos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción , Fosfatidilinositol 3-Quinasas/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
9.
Redox Biol ; 51: 102273, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35255426

RESUMEN

This study investigated the effects of supplementation with a cyanidin- and delphinidin-rich extract (CDRE) on the postprandial dysmetabolism, inflammation, and redox and insulin signaling, triggered by the consumption of a high fat meal (HFM) in healthy individuals. Participants (n = 25) consumed a 1026-kcal HFM simultaneously with either the CDRE providing 320.4 mg of anthocyanins (90% cyanidin and delphinidin) or placebo. Diets were randomly assigned in a double blind, placebo-controlled crossover design. Blood was collected prior to (fasted, time 0), and for 5 h after meal consumption; plasma, serum, and peripheral blood mononuclear cells (PBMC) were isolated. AC metabolites were detected in serum as early as 30 min after CDRE consumption. The CDRE mitigated HFM-induced endotoxemia, reducing increases in plasma LPS and LPS-binding protein. The CDRE also reduced other events associated with HFM-triggered postprandial dysmetabolism including: i) plasma glucose and triglyceride increases; ii) TNFα and NOX4 upregulation in PBMC; and iii) JNK1/2 activation in PBMC. The CDRE did not significantly affect HFM-mediated increases in plasma insulin, GLP-1, GLP-2, GIP, and LDL- and HDL-cholesterol, and IKK phosphorylation in PBMC. In summary, dietary AC, i.e. cyanidin and delphinidin, exerted beneficial actions against unhealthy diets by modulating the associated postprandial dysmetabolism, endotoxemia, alterations of glycemia and lipidemia, and redox and insulin signaling.


Asunto(s)
Antocianinas , Endotoxemia , Antocianinas/farmacología , Antocianinas/uso terapéutico , Glucemia/metabolismo , Estudios Cruzados , Dieta Alta en Grasa/efectos adversos , Endotoxemia/metabolismo , Voluntarios Sanos , Humanos , Insulina , Leucocitos Mononucleares/metabolismo
10.
Mol Nutr Food Res ; 66(21): e2101033, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35182412

RESUMEN

SCOPE: This work studies the capacity of curcumin to inhibit tumor necrosis alpha (TNFα)-induced inflammation, oxidative stress, and loss of intestinal barrier integrity, characterizing the underlying mechanisms. METHODS AND RESULTS: Caco-2 cell monolayers are incubated with TNFα (10 ng mL-1 ), in the absence or presence of curcumin. TNFα causes an increase in interleukin (IL)-6 and IL-8 release, which is inhibited by curcumin in a dose-dependent manner (half-maximal inhibitory concentration (IC50 ) = 3.4 µM for IL-6). Moreover, TNFα leads to: i) increased intercellular adhesion molecule 1 (ICAM-1) and NLRP3 inflammasome expression; ii) increased cell monolayer permeability and decreased levels of tight junction proteins; iii) increased cellular and mitochondrial oxidant production; iv) decreased mitochondrial membrane potential and complex I-III activity; v) activation of redox-sensitive pathways, i.e., nuclear factor-kappa B (NF-κB), extracellular signal-regulated kinase 1/2 (ERK1/2), and c-Jun N-terminal kinases (JNK); and vi) increased myosin light-chain kinase (MLCK) expression and phosphorylation levels of myosin light-chain protein MLC. Curcumin (2-8 µM) inhibits all these TNFα-triggered undesirable outcomes, mostly showing dose-dependent effects. CONCLUSION: The inhibition of NF-κB, ERK1/2, and JNK activation could be in part involved in the capacity of curcumin to mitigate intestinal inflammation, oxidant production, activation of redox-sensitive pathways, and prevention of monolayer permeabilization. These results support an action of dietary curcumin in sustaining gastrointestinal tract physiology.


Asunto(s)
Curcumina , FN-kappa B , Humanos , Células CACO-2 , FN-kappa B/metabolismo , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Curcumina/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Oxidantes , Inflamación
11.
Food Funct ; 13(2): 781-794, 2022 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-34981106

RESUMEN

Consumption of high fat diets (HFD) and the associated metabolic endotoxemia can initiate liver inflammation and lipid deposition that with time can progress to non-alcoholic fatty liver disease (NAFLD). We previously observed that 14 weeks supplementation with the anthocyanidins cyanidin and delphinidin mitigated HFD-induced metabolic endotoxemia and liver insulin resistance, steatosis, inflammation and oxidative stress. This work investigated if a 4-week supplementation of mice with a cyanidin- and delphinidin-rich extract (CDRE) could mitigate or reverse HFD (60% calories from lard fat)-induced liver steatosis and inflammation. After a first 4-weeks period on the HFD, mice showed increased endotoxemia and activation of liver proinflammatory signaling cascades. Supplementation with CDRE between weeks 4 and 8 did not mitigate liver steatosis or the altered lipid and glucose plasma levels. However, CDRE supplementation reverted HFD-induced metabolic endotoxemia, in parallel with the mitigation of the overexpression of hepatic TLR2 and TLR4, and of the activation of: (i) NF-κB, (ii) AP-1 and upstream mitogen-activated kinases p38 and ERK1/2, and (iii) HIF-1. Thus, even a short-term consumption of cyanidin and delphinidin could help mitigate the adverse consequences, i.e. metabolic endotoxemia and associated liver inflammation, triggered by the regular consumption of diets rich in fat.


Asunto(s)
Antocianinas/farmacología , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Dieta Alta en Grasa/efectos adversos , Endotoxemia/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Alimentación Animal , Animales , Suplementos Dietéticos , Endotoxemia/inducido químicamente , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/inducido químicamente , Ratones , FN-kappa B , Estrés Oxidativo , Transducción de Señal , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
12.
Dig Dis Sci ; 67(7): 3006-3016, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-34156590

RESUMEN

BACKGROUND: Deregulation of immune response and oxidative stress contribute to nonalcoholic fatty liver disease (NAFLD) pathogenesis. Resistin is a physiological modulator of inflammation and redox homeostasis of different cell types. Increased resistin serum concentration and the direct association between resistin hepatic expression and NAFLD severity suggest that resistin participates in NAFLD pathogenesis. AIMS: To evaluate resistin-induced regulation of redox homeostasis in mononuclear leukocytes from NAFLD patients and controls. METHODS: We evaluated basal and resistin-mediated modulation of reactive oxygen species (ROS) and glutathione content by flow cytometry, and antioxidant enzyme activities by spectrophotometry. RESULTS: Peripheral blood mononuclear cells (PBMC) from NAFLD patients showed higher ROS content and glutathione peroxidase activity and lower glutathione content, superoxide dismutase and glutathione reductase activities than control PBMC. Resistin decreased ROS levels and superoxide dismutase activity and increased glutathione reductase and catalase activities in PBMC from controls but not from patients. Resistin decreased glutathione content in PBMC from control and NAFLD patients, with greater effect on patient cells. Basal and resistin-modulated ROS levels were directly associated with obesity-related risk factors for NAFLD. Hepatic myeloid cells and T-lymphocytes from NAFLD patients showed higher basal ROS content than cells from controls. Resistin decreased ROS levels in hepatic T-lymphocytes from controls but not from patients. CONCLUSIONS: Resistin regulates redox homeostasis in mononuclear leukocytes. A decreased response to resistin in leukocytes from NAFLD patients is associated with an impaired redox homeostasis.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Antioxidantes/metabolismo , Glutatión/metabolismo , Glutatión Reductasa/metabolismo , Humanos , Leucocitos Mononucleares/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Estrés Oxidativo , Especies Reactivas de Oxígeno , Resistina/metabolismo , Superóxido Dismutasa/metabolismo
13.
Pflugers Arch ; 474(1): 99-115, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34812946

RESUMEN

This review summarizes experimental evidence on the beneficial effects of ( -)-epicatechin (EC) attenuating major cardiometabolic risk factors, i.e., dyslipidemias, obesity (adipose tissue dysfunction), hyperglycemia (insulin resistance), and hypertension (endothelial dysfunction). Studies in humans are revised and complemented with experiments in animal models, and cultured cells, aiming to understand the molecular mechanisms involved in EC-mediated effects. Firstly, an assessment of EC metabolism gives relevance to both conjugated-EC metabolites product of host metabolism and microbiota-derived species. Integration and analysis of results stress the maintenance of redox homeostasis and mitigation of inflammation as relevant processes associated with cardiometabolic diseases. In these processes, EC appears having significant effects regulating NADPH oxidase (NOX)-dependent oxidant production, nitric oxide (NO) production, and energy homeostasis (mitochondrial biogenesis and function). The potential participation of cell membranes and membrane-bound receptors is also discussed in terms of direct molecular action of EC and EC metabolites reaching cells and tissues.


Asunto(s)
Factores de Riesgo Cardiometabólico , Catequina/farmacología , Animales , Catequina/química , Catequina/metabolismo , Catequina/uso terapéutico , Dislipidemias/tratamiento farmacológico , Humanos , Hiperglucemia/tratamiento farmacológico , Hipertensión/tratamiento farmacológico , Obesidad/tratamiento farmacológico
14.
Redox Biol ; 42: 101914, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33750648

RESUMEN

Identification of the links among flavonoid consumption, mitigation of oxidative stress and improvement of disease in humans has significantly advanced in the last decades. This review used (-)-epicatechin (EC) as an example of dietary flavonoids, and inflammation, endothelial dysfunction/hypertension and insulin resistance/diabetes as paradigms of human disease. In these pathologies, oxidative stress is part of their development and/or their perpetuation. Evidence from both, rodent studies and characterization of mechanisms in cell cultures are encouraging and mostly support indirect antioxidant actions of EC and EC metabolites in endothelial dysfunction and insulin resistance. Human studies also show beneficial effects of EC on these pathologies based on biomarkers of disease. However, there is limited available information on oxidative stress biomarkers and flavonoid consumption to allow establishing conclusive associations. The evolving discovery of metabolites that could serve as reliable markers of intake of specific flavonoids constitutes a powerful tool to link flavonoid consumption to disease and prevention of oxidative stress in human populations.


Asunto(s)
Catequina , Flavonoides , Antioxidantes , Biomarcadores , Humanos , Modelos Teóricos , Estrés Oxidativo
15.
Food Funct ; 11(10): 8405-8406, 2020 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-32969452
16.
Food Funct ; 11(7): 5944-5954, 2020 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-32613983

RESUMEN

High adipose tissue (AT) accumulation in the body increases the risk for many metabolic and chronic diseases. This work investigated the capacity of the flavonoid (-)-epicatechin to prevent undesirable modifications of AT in mice fed a high-fat diet. Studies were focused on thoracic aorta perivascular AT (taPVAT), which is involved in the control of blood vessel tone, among other functions. Male C57BL/6J mice were fed for 15 weeks a high-fat diet with or without added (-)-epicatechin (20 mg per kg body weight per d). In high-fat diet fed mice, (-)-epicatechin supplementation: (i) prevented the expansion of taPVAT, (ii) attenuated the whitening of taPVAT (according to the adipocyte morphology, diameter, and uncoupling-protein 1 (UCP-1) levels) and (iii) blunted the increase in plasma glucose and cholesterol. The observed taPVAT modifications were not associated with alterations in the aorta wall thickness, aorta tumor necrosis factor-alpha (TNF-α) and NADPH-oxidase 2 (NOX2) expression, and endothelial nitric oxide synthase (eNOS) phosphorylation levels. In summary, our results indicate (-)-epicatechin as a relevant bioactive protecting from the slow and silent development of metabolic and chronic diseases as they are associated with excessive fat intake.


Asunto(s)
Tejido Adiposo/patología , Aorta Torácica/patología , Catequina/farmacología , Dieta Alta en Grasa/efectos adversos , Grasas de la Dieta/efectos adversos , Suplementos Dietéticos , Extractos Vegetales/farmacología , Adipocitos/metabolismo , Adipocitos/patología , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Tejido Adiposo Blanco , Animales , Aorta Torácica/metabolismo , Glucemia/metabolismo , Catequina/uso terapéutico , Colesterol/sangre , Grasas de la Dieta/administración & dosificación , Masculino , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/patología , Enfermedades Metabólicas/prevención & control , Ratones Endogámicos C57BL , NADPH Oxidasa 2/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Obesidad/metabolismo , Obesidad/prevención & control , Extractos Vegetales/uso terapéutico , Factor de Necrosis Tumoral alfa/metabolismo , Proteína Desacopladora 1/metabolismo
17.
Free Radic Biol Med ; 152: 776-786, 2020 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-31981623

RESUMEN

Chronic intestinal inflammation involves a cycle of oxidative stress, activation of redox sensitive transcription factors, and barrier permeabilization. The latter can lead to systemic inflammation and its associated co-morbidities. Diet can play a major role in the modulation of intestinal inflammation. Among plant bioactives, ellagic acid (EA) was reported to inhibit inflammatory bowel disease in animal models. This work investigated the mechanisms by which EA inhibits tumor necrosis factor alpha (TNFα)-induced inflammation, oxidative stress, and loss of barrier integrity. Caco-2 cells differentiated into an intestinal epithelial cell monolayer were incubated with TNFα (10 ng/ml), in the presence of different EA concentrations. TNFα triggered interleukin (IL) 6 and 8 release into the medium, which was inhibited by EA in a dose-dependent manner (IC50 = 17.3 µM for IL-6). TNFα also led to: i) increased ICAM-1 and NLRP3 expression; ii) loss of epithelial barrier function; iii) increased oxidant production from NOX and mitochondrial origin; iv) NF-κB and ERK1/2 activation; and v) increased MLCK gene expression and MLC phosphorylation. EA (10-40 µM) inhibited all these adverse effects of TNFα. EA mainly acted through NF-κB and ERK1/2 inhibition, breaking the cycle of inflammation, oxidative stress, redox-sensitive pathway (e.g. NF-κB, ERK1/2) activation and intestinal permeabilization. This suggests that consumption of EA, via foods or supplements, may afford a strategy to mitigate intestinal inflammation and its associated co-morbidities.


Asunto(s)
Ácido Elágico , Mucosa Intestinal , Animales , Células CACO-2 , Ácido Elágico/farmacología , Humanos , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Quinasa de Cadena Ligera de Miosina , FN-kappa B/genética , Uniones Estrechas , Factor de Necrosis Tumoral alfa/genética
18.
Food Funct ; 11(1): 318-327, 2020 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-31808777

RESUMEN

The aim of this work was to evaluate the protective effects of (-)-epicatechin on the kidneys of NO-deprived rats. Male Sprague Dawley rats were divided into three groups: control (C), receiving water and standard diet; l-NAME (L), receiving a solution of N(ω)-nitro-l-arginine methyl ester (l-NAME) (360 mg l-1 in water) as a beverage and standard diet; and l-NAME-(-)-epicatechin (LE), receiving l-NAME solution as a beverage and standard diet supplemented with (-)-epicatechin (4 g kg-1 diet). The L-group showed altered kidney function parameters, evaluated based on plasma urea and creatinine. In parallel, kidney oxidative stress markers, i.e. superoxide anion production, malondialdehyde content, and 3-nitrotyrosine protein adducts, were significantly increased in the L group. In addition, l-NAME treatment induced modifications in kidney NO bioavailability determinants: increased expression of NOX subunits (p47phox, gp91phox, NOXO1, and NOX4) and lowered NOS activity. (-)-Epicatechin administration restored kidney function parameters, oxidative stress markers, expression of p47phox, gp91phox, and NOX4 and NOS activity to control values. These results indicate that (-)-epicatechin can mitigate NO-mediated impairment of kidney function, in part due to its capacity to modulate NOXs, NOSs, and consequently oxidative stress, and NO bioavailability.


Asunto(s)
Catequina/farmacología , Riñón/efectos de los fármacos , NG-Nitroarginina Metil Éster/efectos adversos , Estrés Oxidativo , Sustancias Protectoras/farmacología , Animales , Masculino , Malondialdehído/metabolismo , NADPH Oxidasas/metabolismo , Óxido Nítrico/análisis , Ratas , Ratas Sprague-Dawley , Superóxidos/metabolismo
19.
Redox Biol ; 26: 101269, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31330482

RESUMEN

The gastrointestinal (GI) tract can play a critical role in the development of pathologies associated with overeating, overweight and obesity. We previously observed that supplementation with anthocyanins (AC) (particularly glycosides of cyanidin and delphinidin) mitigated high fat diet (HFD)-induced development of obesity, dyslipidemia, insulin resistance and steatosis in C57BL/6J mice. This paper investigated whether these beneficial effects could be related to AC capacity to sustain intestinal monolayer integrity, prevent endotoxemia, and HFD-associated dysbiosis. The involvement of redox-related mechanisms were further investigated in Caco-2 cell monolayers. Consumption of a HFD for 14 weeks caused intestinal permeabilization and endotoxemia, which were associated with a decreased ileum expression of tight junction (TJ) proteins (occludin, ZO-1 and claudin-1), increased expression of NADPH oxidase (NOX1 and NOX4) and NOS2 and oxidative stress, and activation of redox sensitive signals (NF-κB and ERK1/2) that regulate TJ dynamics. AC supplementation mitigated all these events and increased GLP-2 levels, the intestinal hormone that upregulates TJ protein expression. AC also prevented, in vitro, tumor necrosis factor alpha-induced Caco-2 monolayer permeabilization, NOX1/4 upregulation, oxidative stress, and NF-κB and ERK activation. HFD-induced obesity in mice caused dysbiosis and affected the levels and secretion of MUC2, a mucin that participates in intestinal cell barrier protection and immune response. AC supplementation restored microbiota composition and MUC2 levels and distribution in HFD-fed mice. Thus, AC, particularly delphinidin and cyanidin, can preserve GI physiology in HFD-induced obesity in part through redox-regulated mechanisms. This can in part explain AC capacity to mitigate pathologies, i.e. insulin resistance and steatosis, associated with HFD-associated obesity.


Asunto(s)
Antocianinas/farmacología , Dieta Alta en Grasa/efectos adversos , Microbioma Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/metabolismo , Oxidación-Reducción , Sustancias Protectoras/farmacología , Células CACO-2 , Disbiosis , Endotoxemia/tratamiento farmacológico , Endotoxemia/etiología , Endotoxemia/metabolismo , Células Caliciformes/metabolismo , Humanos , Mucina 2/genética , Mucina 2/metabolismo , FN-kappa B/metabolismo , Estrés Oxidativo/efectos de los fármacos , Permeabilidad/efectos de los fármacos , Transducción de Señal
20.
Food Funct ; 10(2): 514-528, 2019 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-30746536

RESUMEN

Although deficiencies in polyphenol intake do not result in specific deficiency diseases, adequate intake of polyphenols could confer health benefits, especially with regard to chronic diseases. Tea, cocoa, fruits, and berries, as well as vegetables, are rich in polyphenols. Flavan-3-ols from cocoa have been found to be associated with a reduced risk of stroke, myocardial infarction, and diabetes, as well as improvements in lipids, endothelial-dependent blood flow and blood pressure, insulin resistance, and systemic inflammation. The flavonoid quercetin and the stilbene resveratrol have also been associated with cardiometabolic health. Although polyphenols have been associated with improved cerebral blood flow, evidence of an impact on cognition is more limited. The ability of dietary polyphenols to produce clinical effects may be due, at least in part, to a bi-directional relationship with the gut microbiota. Polyphenols can impact the composition of the gut microbiota (which are independently associated with health benefits), and gut bacteria metabolize polyphenols into bioactive compounds that produce clinical benefits. Another critical interaction is that of polyphenols with other phytochemicals, which could be relevant to interpreting the health parameter effects of polyphenols assayed as purified extracts, whole foods, or whole food extracts.


Asunto(s)
Análisis de los Alimentos , Alimentos Funcionales/análisis , Polifenoles/farmacología , Flavonoides/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Resistencia a la Insulina , Polifenoles/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...