Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Gut Microbes ; 15(1): 2221095, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37305942

RESUMEN

Impacts of dietary fiber on intestinal inflammation are complex, but some specific semi-purified fibers, particularly psyllium, can protect humans and rodents against colitis. Mechanisms underlying such protection are not fully understood but may involve activation of the FXR bile acid receptor. Obesity and its associated consequences, referred to as metabolic syndrome, are associated with, and promoted by, low-grade inflammation in a variety of tissues including the intestine. Hence, we examined whether psyllium might ameliorate the low-grade intestinal inflammation that occurs in diet-induced obesity and, moreover, the extent to which it might ameliorate adiposity and/or dysglycemia in this disease model. We observed that enriching a high-fat diet with psyllium provided strong protection against the low-grade gut inflammation and metabolic consequences that were otherwise induced by the obesogenic diet. Such protection was fully maintained in FXR-deficient mice, indicating that distinct mechanisms mediate psyllium's protection against colitis and metabolic syndrome. Nor did psyllium's protection associate with, or require, fermentation or IL-22 production, both of which are key mediators of beneficial impacts of some other dietary fibers. Psyllium's beneficial impacts were not evident in germfree mice but were observed in Altered Schaedler Flora mice, in which psyllium modestly altered relative and absolute abundance of the small number of taxa present in these gnotobiotic mice. Thus, psyllium protects mice against diet-induced obesity/metabolic syndrome by a mechanism independent of FXR and fermentation but nonetheless requires the presence of at least a minimal microbiota.


Asunto(s)
Colitis , Microbioma Gastrointestinal , Síndrome Metabólico , Psyllium , Humanos , Animales , Ratones , Síndrome Metabólico/prevención & control , Dieta Occidental , Obesidad/prevención & control , Fibras de la Dieta , Inflamación
2.
Cell Mol Gastroenterol Hepatol ; 15(6): 1421-1442, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36828279

RESUMEN

BACKGROUND & AIMS: Fiber-rich foods promote health, but mechanisms by which they do so remain poorly defined. Screening fiber types, in mice, revealed psyllium had unique ability to ameliorate 2 chronic inflammatory states, namely, metabolic syndrome and colitis. We sought to determine the mechanism of action of the latter. METHODS: Mice were fed grain-based chow, which is naturally rich in fiber or compositionally defined diets enriched with semi-purified fibers. Mice were studied basally and in models of chemical-induced and T-cell transfer colitis. RESULTS: Relative to all diets tested, mice consuming psyllium-enriched compositionally defined diets were markedly protected against both dextran sulfate sodium- and T-cell transfer-induced colitis, as revealed by clinical-type, histopathologic, morphologic, and immunologic parameters. Such protection associated with stark basal changes in the gut microbiome but was independent of fermentation and, moreover, maintained in mice harboring a minimal microbiota (ie, Altered Schaedler Flora). Transcriptomic analysis revealed psyllium induced expression of genes mediating bile acids (BA) secretion, suggesting that psyllium's known ability to bind BA might contribute to its ability to prevent colitis. As expected, psyllium resulted in elevated level of fecal BA, reflecting their removal from enterohepatic circulation but, in stark contrast to the BA sequestrant cholestyramine, increased serum BA levels. Moreover, the use of BA mimetics that activate the farnesoid X receptor (FXR), as well as the use of FXR-knockout mice, suggested that activation of FXR plays a central role in psyllium's protection against colitis. CONCLUSIONS: Psyllium protects against colitis via altering BA metabolism resulting in activation of FXR, which suppresses pro-inflammatory signaling.


Asunto(s)
Colitis , Psyllium , Ratones , Animales , Psyllium/efectos adversos , Ácidos y Sales Biliares , Promoción de la Salud , Colitis/inducido químicamente , Colitis/prevención & control , Colitis/metabolismo , Inflamación , Ratones Noqueados
3.
Curr Dev Nutr ; 6(10): nzac105, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36204330

RESUMEN

Background: Purified diets (PDs) contain refined ingredients with one main nutrient, allowing for greater control relative to grain-based diets (GBDs), which contain unrefined grains and animal byproducts. Traditional PDs like the American Institute of Nutrition (AIN)-76A (76A) and AIN-93G (93G) can negatively impact metabolic and gut health when fed long term, in part due to lower total fiber, no soluble fiber, and higher sucrose content. Objective: Two studies were conducted to determine how PDs with reduced sucrose and increased fiber (soluble and insoluble) influence metabolic and gut health in mice compared with traditional AIN PDs or GBDs. Methods: In study 1, C57Bl/6N mice (n = 75) consumed a GBD [LabDiet 5002 (5002)], 76A, 93G, or 2 PDs with reduced sucrose and higher fiber for 88 d. Body composition and metabolic parameters were assessed. In study 2, C57Bl/6N mice (n = 54) consumed either 2 GBDs (LabDiet 5001 or 5002) or PDs with different types/levels of fiber for 14 d. Microbiome alterations and predicted functional metagenomic changes were measured. Results: The PD with 75 g cellulose and 25 g inulin per 4084 kcals marginally influenced body weight and adiposity, but improved glucose tolerance relative to 93G (P = 0.0131) and 76A (P = 0.0014). Cecal and colonic weights were lower in mice fed cellulose-based PDs compared with those fed GBDs and soluble-fiber PDs. Soluble-fiber PDs reduced alpha diversity and showed similar beta diversity, which differed from cellulose-based PDs and GBDs. Certain genera associated with improved gut health such as Bifidobacteria and Akkermansia were significantly elevated by soluble-fiber PDs (P ≤ 0.01). Metabolic pathways related to carbohydrate and fatty acid metabolism were affected by PDs. Conclusions: PDs formulated with lower sucrose and increased fiber content, particularly soluble fiber, blunted elevations in metabolic parameters and favorably impacted the microbiota and metagenome in C57BL/6N mice.

4.
Nutrients ; 13(12)2021 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-34959832

RESUMEN

Refined foods are commonly depleted in certain bioactive components that are abundant in 'natural' (plant) foods. Identification and addition of these 'missing' bioactives in the diet is, therefore, necessary to counteract the deleterious impact of convenience food. In this study, multiomics approaches were employed to assess the addition of the popular supplementary soluble dietary fibers inulin and psyllium, both in isolation and in combination with a refined animal feed. A 16S rRNA sequencing and 1H NMR metabolomic investigation revealed that, whilst inulin mediated an increase in Bifidobacteria, psyllium elicited a broader microbial shift, with Parasutterella and Akkermansia being increased and Enterorhabdus and Odoribacter decreased. Interestingly, the combination diet benefited from both inulin and psyllium related microbial changes. Psyllium mediated microbial changes correlated with a reduction of glucose (R -0.67, -0.73, respectively, p < 0.05) and type 2 diabetes associated metabolites: 3-methyl-2-oxovaleric acid (R -0.72, -0.78, respectively, p < 0.05), and citrulline (R -0.77, -0.71, respectively, p < 0.05). This was in line with intestinal and hepatic carbohydrate response (e.g., Slc2a2, Slc2a5, Khk and Fbp1) and hepatic lipogenesis (e.g., Srebf1 and Fasn), which were significantly reduced under psyllium addition. Although established in the liver, the intestinal response associated with psyllium was absent in the combination diet, placing greater significance upon the established microbial, and subsequent metabolomic, shift. Our results therefore highlight the heterogeneity that exists between distinct dietary fibers in the context of carbohydrate uptake and metabolism, and supports psyllium containing combination diets, for their ability to negate the impact of a refined diet.


Asunto(s)
Fibras de la Dieta/farmacología , Suplementos Dietéticos , Inulina/farmacología , Psyllium/farmacología , Alimentación Animal , Animales , Dieta/métodos , Comida Rápida , Microbioma Gastrointestinal/efectos de los fármacos , Glucosa/metabolismo , Intestinos/metabolismo , Lipogénesis/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fitoquímicos/farmacología , ARN Ribosómico 16S/análisis
5.
Cell Mol Gastroenterol Hepatol ; 12(3): 983-1000, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33940221

RESUMEN

BACKGROUND & AIMS: Nourishment of gut microbiota via consumption of fermentable fiber promotes gut health and guards against metabolic syndrome. In contrast, how dietary fiber impacts type 1 diabetes is less clear. METHODS: To examine impact of dietary fibers on development of type 1 diabetes in the streptozotocin (STZ)-induced and spontaneous non-obese diabetes (NOD) models, mice were fed grain-based chow (GBC) or compositionally defined diets enriched with a fermentable fiber (inulin) or an insoluble fiber (cellulose). Spontaneous (NOD mice) or STZ-induced (wild-type mice) diabetes was monitored. RESULTS: Relative to GBC, low-fiber diets exacerbated STZ-induced diabetes, whereas diets enriched with inulin, but not cellulose, strongly protected against or treated it. Inulin's restoration of glycemic control prevented loss of adipose depots, while reducing food and water consumption. Inulin normalized pancreatic function and markedly enhanced insulin sensitivity. Such amelioration of diabetes was associated with alterations in gut microbiota composition and was eliminated by antibiotic administration. Pharmacologic blockade of fermentation reduced inulin's beneficial impact on glycemic control, indicating a role for short-chain fatty acids (SCFA). Furthermore, inulin's microbiota-dependent anti-diabetic effect associated with SCFA-independent restoration of interleukin 22, which was necessary and sufficient to ameliorate STZ-induced diabetes. Inulin-enriched diets significantly delayed diabetes in NOD mice. CONCLUSIONS: Fermentable fiber confers microbiota-dependent increases in SCFA and interleukin 22 that, together, may have potential to prevent and/or treat type 1 diabetes.


Asunto(s)
Bacterias/clasificación , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Fibras de la Dieta/administración & dosificación , Ácidos Grasos Volátiles/metabolismo , Interleucinas/metabolismo , Inulina/administración & dosificación , Animales , Bacterias/efectos de los fármacos , Bacterias/genética , Bacterias/aislamiento & purificación , Diabetes Mellitus Tipo 1/inducido químicamente , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/microbiología , Fibras de la Dieta/farmacología , Modelos Animales de Enfermedad , Microbioma Gastrointestinal/efectos de los fármacos , Hemoglobina Glucada/metabolismo , Inulina/farmacología , Masculino , Ratones , Ratones Endogámicos NOD , Tamaño de los Órganos/efectos de los fármacos , Páncreas/efectos de los fármacos , Páncreas/fisiopatología , Estreptozocina/efectos adversos , Resultado del Tratamiento , Interleucina-22
6.
Curr Dev Nutr ; 5(12): nzab138, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34993389

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is intricately linked to metabolic disease (including obesity, glucose intolerance, and insulin resistance) and encompasses a spectrum of disorders including steatosis, nonalcoholic steatohepatitis (NASH), and fibrosis. Rodents consuming high-fat (HF; ∼40 kcal% fat including fats containing higher concentrations of saturated and trans fats), high-fructose (HFr), and high-cholesterol (HC) diets display many clinically relevant characteristics of NASH, along with other metabolic disorders. C57BL/6 mice are the most commonly used animal model because they can develop significant metabolic disorders including severe NASH with fibrosis after months of feeding, but other models also are susceptible. The significant number of diets that contain these different factors (i.e., HF, HFr, and HC), either alone or in combination, makes the choice of diet difficult. This methodology review describes the efficacy of these nutrient manipulations on the NAFLD phenotype in mice, rats, guinea pigs, hamsters, and nonhuman primates.

7.
Mol Nutr Food Res ; 64(17): e2000162, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32656952

RESUMEN

SCOPE: Previous studies have suggested that diets rich in omega-3 and low in omega-6 long-chain polyunsaturated fatty acids (PUFAs) can limit the development of metabolic syndrome (MetS). Transgenic soybeans yielding oils enriched for omega-3 PUFAs represent a new and readily-available option for incorporating omega-3 PUFAs into diets to provide health benefits. METHODS AND RESULTS: Transgenic soybean oils, enriched for either stearidonic acid (SDA) or eicosapentaenoic acid (EPA), are incorporated into diets to test their effects on limiting the development of MetS in a mouse model of diet-induced obesity. Supplementation with SDA- but not EPA-enriched oils improved features of MetS compared to feeding a control wild-type oil. Because previous studies have linked the gut microorganism Akkermansia muciniphila to the metabolic effects of feeding omega-3 PUFAs, the causal contribution of A. muciniphila to mediating the metabolic benefits provided by SDA-enriched diets is investigated. Although A. muciniphila is not required for SDA-induced metabolic improvements, this microorganism does modulate levels of saturated and mono-unsaturated fatty acids in host adipose tissues. CONCLUSION: Together, these findings support the utilization of SDA-enriched diets to modulate weight gain, glucose metabolism, and fatty acid profiles of liver and adipose tissue.


Asunto(s)
Ácidos Grasos Omega-3/farmacología , Glucosa/metabolismo , Obesidad/dietoterapia , Aceite de Soja/farmacología , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Akkermansia/efectos de los fármacos , Akkermansia/fisiología , Animales , Dieta Alta en Grasa/efectos adversos , Suplementos Dietéticos , Ácido Eicosapentaenoico/farmacología , Ácidos Grasos Insaturados/farmacocinética , Alimentos Fortificados , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/fisiología , Masculino , Ratones Endogámicos C57BL , Obesidad/metabolismo , Obesidad/microbiología , Plantas Modificadas Genéticamente , Aceite de Soja/química , Aceite de Soja/genética , Aumento de Peso/efectos de los fármacos
8.
Curr Dev Nutr ; 4(6): nzaa078, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32494762

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is a complex spectrum of disorders ranging from simple benign steatosis to more aggressive forms of nonalcoholic steatohepatitis (NASH) and fibrosis. Although not every patient with NAFLD/NASH develops liver complications, if left untreated it may eventually lead to cirrhosis and hepatocellular carcinoma. Purified diets formulated with specific nutritional components can drive the entire spectrum of NAFLD in rodent models. Although they may not perfectly replicate the clinical and histological features of human NAFLD, they provide a model to gain further understanding of disease progression in humans. Owing to the growing demand of diets for NAFLD research, and for our further understanding of how manipulation of dietary components can alter disease development, we outlined several commonly used dietary approaches for rodent models, including mice, rats, and hamsters, time frames required for disease development and whether other metabolic diseases commonly associated with NAFLD in humans occur.

9.
Curr Dev Nutr ; 4(4): nzaa031, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32258990

RESUMEN

The reproducibility of experimental data is challenged by many factors in both clinical and preclinical research. In preclinical studies, several factors may be responsible, and diet is one variable that is commonly overlooked, especially by those not trained in nutrition. In particular, grain-based diets contain complex ingredients, each of which can provide multiple nutrients, non-nutrients, and contaminants, which may vary from batch to batch. Thus, even when choosing the same grain-based diet used in the past by others, its composition will likely differ. In contrast, purified diets contain refined ingredients that offer the ability to control the composition much more closely and maintain consistency from one batch to the next, while minimizing the presence of non-nutrients and contaminants. In this article, we provide several different examples or scenarios showing how the diet choice can alter data interpretation, potentially affecting reproducibility and knowledge gained within any given field of study.

10.
Curr Protoc Toxicol ; 77(1): e55, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30028909

RESUMEN

Poor diet reporting and improperly controlling laboratory animal diet continues to reduce our ability to interpret data effectively in animal studies. In order to make the best use of our resources and improve research transparency, proper reporting methods that include a diet design are essential to improving our understanding of the links between gut health and metabolic disease onset. This unit will focus on the importance of diet choice in laboratory animal studies, specifically as it relates to gut health, microbiome, and metabolic disease development. The two most commonly used diet types, grain-based (GB) diets, and purified ingredient diets, will each be described, with particular emphasis on their differences in dietary fiber. A further description of how these diet types and fiber can affect gut morphology and microbiota will be provided as well as how purified ingredient diets may be improved upon. © 2018 by John Wiley & Sons, Inc.

11.
Cell ; 174(2): 271-284.e14, 2018 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-29887373

RESUMEN

The small intestinal tuft cell-ILC2 circuit mediates epithelial responses to intestinal helminths and protists by tuft cell chemosensory-like sensing and IL-25-mediated activation of lamina propria ILC2s. Small intestine ILC2s constitutively express the IL-25 receptor, which is negatively regulated by A20 (Tnfaip3). A20 deficiency in ILC2s spontaneously triggers the circuit and, unexpectedly, promotes adaptive small-intestinal lengthening and remodeling. Circuit activation occurs upon weaning and is enabled by dietary polysaccharides that render mice permissive for Tritrichomonas colonization, resulting in luminal accumulation of acetate and succinate, metabolites of the protist hydrogenosome. Tuft cells express GPR91, the succinate receptor, and dietary succinate, but not acetate, activates ILC2s via a tuft-, TRPM5-, and IL-25-dependent pathway. Also induced by parasitic helminths, circuit activation and small intestinal remodeling impairs infestation by new helminths, consistent with the phenomenon of concomitant immunity. We describe a metabolic sensing circuit that may have evolved to facilitate mutualistic responses to luminal pathosymbionts.


Asunto(s)
Intestino Delgado/fisiología , Tritrichomonas/metabolismo , Acetatos/metabolismo , Animales , Fibras de la Dieta/metabolismo , Metabolismo Energético , Células Epiteliales/citología , Células Epiteliales/metabolismo , Células Epiteliales/parasitología , Interleucinas/genética , Interleucinas/metabolismo , Mucosa Intestinal/citología , Intestino Delgado/microbiología , Intestino Delgado/parasitología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microbiota , Plásmidos/genética , Plásmidos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Interleucina/metabolismo , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/metabolismo , Ácido Succínico/metabolismo , Canales Catiónicos TRPM/metabolismo , Tritrichomonas/crecimiento & desarrollo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo
12.
Nutr Metab (Lond) ; 15: 23, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29611545

RESUMEN

[This corrects the article DOI: 10.1186/s12986-018-0243-5.].

13.
Nutr Metab (Lond) ; 15: 3, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29371873

RESUMEN

Diets used to induce metabolic disease are generally high in fat and refined carbohydrates and importantly, are usually made with refined, purified ingredients. However, researchers will often use a low fat grain-based (GB) diet containing unrefined ingredients as the control diet. Such a comparison between two completely different diet types makes it impossible to draw conclusions regarding the phenotypic differences driven by diet. While many compositional differences can account for this, one major difference that could have the greatest impact between GB and purified diets is the fiber content, both in terms of the level and composition. We will review recent data showing how fiber differences between GB diets and purified diets can significantly influence gut health and microbiota, which itself can affect metabolic disease development. Researchers need to consider the control diet carefully in order to make the best use of precious experimental resources.

14.
Genetics ; 208(1): 399-417, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29158425

RESUMEN

The incidence of diet-induced metabolic disease has soared over the last half-century, despite national efforts to improve health through universal dietary recommendations. Studies comparing dietary patterns of populations with health outcomes have historically provided the basis for healthy diet recommendations. However, evidence that population-level diet responses are reliable indicators of responses across individuals is lacking. This study investigated how genetic differences influence health responses to several popular diets in mice, which are similar to humans in genetic composition and the propensity to develop metabolic disease, but enable precise genetic and environmental control. We designed four human-comparable mouse diets that are representative of those eaten by historical human populations. Across four genetically distinct inbred mouse strains, we compared the American diet's impact on metabolic health to three alternative diets (Mediterranean, Japanese, and Maasai/ketogenic). Furthermore, we investigated metabolomic and epigenetic alterations associated with diet response. Health effects of the diets were highly dependent on genetic background, demonstrating that individualized diet strategies improve health outcomes in mice. If similar genetic-dependent diet responses exist in humans, then a personalized, or "precision dietetics," approach to dietary recommendations may yield better health outcomes than the traditional one-size-fits-all approach.


Asunto(s)
Dietética , Metabolismo Energético , Estado de Salud , Animales , Composición Corporal , Dieta , Modelos Animales de Enfermedad , Glucosa/metabolismo , Humanos , Hígado/metabolismo , Enfermedades Metabólicas/etiología , Enfermedades Metabólicas/metabolismo , Ratones , Fenotipo
15.
Cell Host Microbe ; 23(1): 41-53.e4, 2018 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-29276170

RESUMEN

Dietary supplementation with fermentable fiber suppresses adiposity and the associated parameters of metabolic syndrome. Microbiota-generated fiber-derived short-chain fatty acids (SCFAs) and free fatty acid receptors including GPR43 are thought to mediate these effects. We find that while fermentable (inulin), but not insoluble (cellulose), fiber markedly protected mice against high-fat diet (HFD)-induced metabolic syndrome, the effect was not significantly impaired by either inhibiting SCFA production or genetic ablation of GPR43. Rather, HFD decimates gut microbiota, resulting in loss of enterocyte proliferation, leading to microbiota encroachment, low-grade inflammation (LGI), and metabolic syndrome. Enriching HFD with inulin restored microbiota loads, interleukin-22 (IL-22) production, enterocyte proliferation, and antimicrobial gene expression in a microbiota-dependent manner, as assessed by antibiotic and germ-free approaches. Inulin-induced IL-22 expression, which required innate lymphoid cells, prevented microbiota encroachment and protected against LGI and metabolic syndrome. Thus, fermentable fiber protects against metabolic syndrome by nourishing microbiota to restore IL-22-mediated enterocyte function.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Fibras de la Dieta/uso terapéutico , Ácidos Grasos Volátiles/metabolismo , Interleucinas/metabolismo , Mucosa Intestinal/microbiología , Inulina/uso terapéutico , Receptores Acoplados a Proteínas G/genética , Adiposidad/efectos de los fármacos , Animales , Bifidobacterium/crecimiento & desarrollo , Proliferación Celular/efectos de los fármacos , Suplementos Dietéticos , Células Epiteliales/metabolismo , Microbioma Gastrointestinal/efectos de los fármacos , Proteínas de Homeodominio/genética , Interleucinas/genética , Mucosa Intestinal/citología , Mucosa Intestinal/crecimiento & desarrollo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/prevención & control , Interleucina-22
16.
Inflamm Bowel Dis ; 23(7): 1133-1143, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28590342

RESUMEN

BACKGROUND: Lack of dietary fiber has been suggested to increase the risk of developing various chronic inflammatory diseases, whereas supplementation of diets with fiber might offer an array of health-promoting benefits. Consistent with this theme, we recently reported that in mice, compositionally defined diets that are made with purified ingredients and lack fermentable fiber promote low-grade inflammation and metabolic syndrome, both of which could be ameliorated by supplementation of such diets with the fermentable fiber inulin. METHODS: Herein, we examined if, relative to a grain-based mouse diet (chow), compositionally defined diet consumption would impact development of intestinal inflammation induced by dextran sulfate sodium (DSS) and moreover, whether DSS-induced colitis might also be attenuated by diets supplemented with inulin. RESULTS: Analogous to their promotion of low-grade inflammation, compositionally defined diet of high- and low-fat content with cellulose increased the severity of DSS-induced colitis relative to chow. However, in contrast to the case of low-grade inflammation, addition of inulin, but not the insoluble fiber cellulose, further exacerbated the severity of colitis and its associated clinical manifestations (weight loss and bleeding) in both low- and high-fat diets. CONCLUSIONS: While inulin, and perhaps other fermentable fibers, can ameliorate low-grade inflammation and associated metabolic disease, it also has the potential to exacerbate disease severity in response to inducers of acute colitis.


Asunto(s)
Colitis/patología , Sulfato de Dextran/toxicidad , Dieta con Restricción de Grasas , Dieta Alta en Grasa , Fibras de la Dieta/administración & dosificación , Suplementos Dietéticos , Inflamación/patología , Animales , Colitis/inducido químicamente , Fermentación , Inflamación/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Pérdida de Peso
18.
Am J Physiol Renal Physiol ; 310(9): F812-20, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26841823

RESUMEN

The majority of patients with obesity, insulin resistance, and metabolic syndrome have hypertension, but the mechanisms of hypertension are poorly understood. In these patients, impaired sodium excretion is critical for the genesis of Na(+)-sensitive hypertension, and prior studies have proposed a role for the epithelial Na(+) channel (ENaC) in this syndrome. We characterized high fat-fed mice as a model in which to study the contribution of ENaC-mediated Na(+) reabsorption in obesity and insulin resistance. High fat-fed mice demonstrated impaired Na(+) excretion and elevated blood pressure, which was significantly higher on a high-Na(+) diet compared with low fat-fed control mice. However, high fat-fed mice had no increase in ENaC activity as measured by Na(+) transport across microperfused cortical collecting ducts, electrolyte excretion, or blood pressure. In addition, we found no difference in endogenous urinary aldosterone excretion between groups on a normal or high-Na(+) diet. High fat-fed mice provide a model of metabolic syndrome, recapitulating obesity, insulin resistance, impaired natriuresis, and a Na(+)-sensitive elevation in blood pressure. Surprisingly, in contrast to previous studies, our data demonstrate that high fat feeding of mice impairs natriuresis and produces elevated blood pressure that is independent of ENaC activity and likely caused by increased Na(+) reabsorption upstream of the aldosterone-sensitive distal nephron.


Asunto(s)
Presión Sanguínea/efectos de los fármacos , Canales Epiteliales de Sodio/metabolismo , Resistencia a la Insulina , Obesidad/metabolismo , Sodio/farmacología , Aldosterona/orina , Animales , Ritmo Circadiano , Dieta Alta en Grasa , Túbulos Renales Colectores/efectos de los fármacos , Túbulos Renales Colectores/metabolismo , Masculino , Síndrome Metabólico/metabolismo , Ratones , Ratones Endogámicos C57BL , Natriuresis , Nefronas/efectos de los fármacos , Nefronas/metabolismo , Obesidad/etiología , Sodio/orina , Sodio en la Dieta/efectos adversos
19.
Am J Physiol Gastrointest Liver Physiol ; 309(7): G528-41, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26185332

RESUMEN

Diet-induced obesity is often modeled by comparing mice fed high-fat diet (HFD), which is made from purified ingredients, vs. normal chow diet (NCD), which is a low-fat assemblage of relatively unrefined plant and animal products. The mechanism by which HFD promotes adiposity is complex but thought to involve low-grade inflammation and altered gut microbiota. The goal of this study was to investigate the extent to which HFD-induced adiposity is driven by fat content vs. other factors that differentiate HFD vs. NCD. Mice were fed NCD, HFD, or other compositionally defined diets (CDD), designed to mimic NCD and/or explore the role of HFD components. A range of metabolic parameters reflecting low-grade inflammation and adiposity were assayed. Relative to NCD, HFD, and to a lesser, but, nonetheless, significant extent, CDD induced increased adiposity, indicating both lipid content and other aspects of HFD are obesogenic. Moreover, HFD and CDD induced a rapid and marked loss of cecal and colonic mass. Such CDD-induced effects were not affected by adjusting dietary protein levels/types but could be largely eliminated by exchanging insoluble fiber (cellulose) for soluble fiber (inulin). Replacing cellulose with inulin in HFD also protected mice against decreased intestinal mass, hyperphagia, and increased adiposity. Such beneficial effects of inulin were microbiota dependent, correlated with elevated fecal short-chain fatty acid levels analyzed via (1)H-NMR-based metabolomics and were partially recapitulated by administration of short-chain fatty acid. HFD-induced obesity is strongly promoted by its lack of soluble fiber, which supports microbiota-mediated intestinal tissue homeostasis that prevents inflammation driving obesity and metabolic syndrome.


Asunto(s)
Tejido Adiposo/metabolismo , Adiposidad , Dieta Alta en Grasa/efectos adversos , Fibras de la Dieta/metabolismo , Obesidad/etiología , Animales , Grasas de la Dieta/metabolismo , Microbioma Gastrointestinal , Inflamación , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo , Espectroscopía de Protones por Resonancia Magnética
20.
Iran J Allergy Asthma Immunol ; 13(4): 247-55, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24659160

RESUMEN

There has been considerable inconsistency regarding the potential relationship between dyslipidemia and bone metabolism. The inflammatory stimulation through the receptor activator of the nuclear factor kappa-B ligand (RANKL)/ receptor activator of the nuclear factor kappa-B (RANK)/ osteoprotegerin (OPG) pathway could be the infrastructural mechanism for hypercholesterolemia-induced bone loss.In this study, we investigated the effect of dyslipidemia on RANKL and OPG alongside with pro-inflammatory cytokines. Thirty male C57Bl/6 mice (4 weeks old) were randomized to two purified diet groups (15 animals in each group), high fat, low carbohydrate diet (HFLCD) and its matched low fat, high carbohydrate diet (LFHCD). After 12 weeks of feeding in standard situations, the plasma concentration of lipid profile, interleukin (IL) 1Beta, IL-6, tumor necrosis factor-alpha (TNF-α) and RANKL, OPG, and RANKL: OPG ratio were measured.In the present study, although the body weight significantly increased during 12 weeks in HFLCD and LFHCD groups, there were no significant differences in food intake, food efficiency ratio and weight gain between the two groups. The LFHCD group had significantly higher median RANKL and RANKL/OPG ratio. There was no significant difference in plasma IL-1ß, IL-6 and TNF-α concentration between LFHCD and HFLCD groups.These unexpected findings from LFHCD, that seem to be as a result of its higher carbohydrate proportion in comparison to HFLCD, implicate dietary carbohydrate rather than dietary fat as a more significant nutritional factor contributing to change in RANKL level and RANKL: OPG ratio.


Asunto(s)
Citocinas/sangre , Dieta Alta en Grasa , Carbohidratos de la Dieta/administración & dosificación , Grasas de la Dieta/administración & dosificación , Osteoprotegerina/sangre , Ligando RANK/sangre , Receptor Activador del Factor Nuclear kappa-B/sangre , Animales , Masculino , Ratones , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...