Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Lipid Res ; 56(6): 1110-22, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25833686

RESUMEN

Interleukin 17A (IL-17A) is a proinflammatory cytokine involved in the pathogenesis of chronic inflammatory diseases. In the field of immunometabolism, we have studied the impact of IL-17A on the lipid metabolism of human in vitro-generated monocyte-derived dendritic cells (DCs). Microarrays and lipidomic analysis revealed an intense remodeling of lipid metabolism induced by IL-17A in DCs. IL-17A increased 2-12 times the amounts of phospholipids, cholesterol, triglycerides, and cholesteryl esters in DCs. Palmitic (16:0), stearic (18:0), and oleic (18:ln-9c) acid were the main fatty acid chains present in DCs. They were strongly increased in response to IL-17A while their relative proportion remained unchanged. Capture of extracellular lipids was the major mechanism of lipid droplet accumulation, visualized by electron microscopy and Oil Red O staining. Besides this foamy phenotype, IL-17A induced a mixed macrophage-DC phenotype and expression of the nuclear receptor NR1H3/liver X receptor-α, previously identified in the context of atherosclerosis as the master regulator of cholesterol homeostasis in macrophages. These IL-17A-treated DCs were as competent as untreated DCs to stimulate allogeneic naive T-cell proliferation. Following this first characterization of lipid-rich DCs, we propose to call these IL-17A-dependent cells "foamy DCs" and discuss the possible existence of foamy DCs in atherosclerosis, a metabolic and inflammatory disorder involving IL-17A.


Asunto(s)
Aterosclerosis/metabolismo , Células Dendríticas/metabolismo , Ácidos Grasos/metabolismo , Interleucina-17/metabolismo , Aterosclerosis/inmunología , Aterosclerosis/patología , Diferenciación Celular/genética , Proliferación Celular/genética , Células Dendríticas/inmunología , Células Espumosas/metabolismo , Regulación de la Expresión Génica , Humanos , Interleucina-17/genética , Interleucina-17/inmunología , Gotas Lipídicas/inmunología , Gotas Lipídicas/metabolismo , Metabolismo de los Lípidos/inmunología , Receptores X del Hígado , Macrófagos/inmunología , Macrófagos/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , Receptores Nucleares Huérfanos/biosíntesis , Receptores Nucleares Huérfanos/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo
2.
J Nutr ; 145(8): 1754-62, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26108543

RESUMEN

BACKGROUND: High-fat diets induce intestinal barrier alterations and promote intestinal diseases. Little is known about the effects of long-chain fatty acids (LCFAs) on mucin 2 (MUC2) production by goblet cells, which are crucial for intestinal protection. OBJECTIVE: We investigated the effects of LCFAs on the differentiation of colonic goblet cells, MUC2 expression, and colonic barrier function. METHODS: Upon reaching confluence, human colonic mucus-secreting HT29-MTX cells were stimulated (21 d) with a saturated LCFA (palmitic or stearic acid), a monounsaturated LCFA (oleic acid), or a polyunsaturated LCFA (linoleic, γ-linolenic, α-linolenic, or eicosapentaenoic acid). In addition, rat pups underwent oral administration of oil (palm, rapeseed, or sunflower oil) or water (10 µL/g body weight, postnatal days 10-15). Subsequently, colon goblet cells were studied by Western blotting, reverse transcriptase-quantitative polymerase chain reaction, and immunohistochemistry and colonic transmucosal electrical resistance was measured by using Ussing chambers. RESULTS: In vitro, palmitic acid enhanced MUC2 production (140% of control) and hepatocyte nuclear factor 4α expression, whereas oleic, linoleic, γ-linolenic, α-linolenic, and eicosapentaenoic acids reduced MUC2 expression (at least -50% of control). All unsaturated LCFAs decreased the expression of human atonal homolog 1, a transcription factor controlling goblet cell differentiation (at least -31% vs. control). In vivo, rats fed palm oil had higher palmitic acid concentrations (3-fold) in their colonic contents and increased mucus granule surfaces in their goblet cells (>2-fold) than did all other groups. Palm oil also increased colonic transmucosal electrical resistance (245% of control), yet had no effect on occludin and zonula occludens-1 expression. In contrast, sunflower and rapeseed oils decreased goblet cell number when compared with control (at least -10%) and palm oil (at least -14%) groups. CONCLUSIONS: Palm oil in rat pups and palmitic acid in HT29-MTX cells increase the production of MUC2 and strengthen the intestinal barrier. In contrast, unsaturated LCFAs decrease MUC2 expression. These data should be taken into account in the context of preventive or therapeutic nutritional programs.


Asunto(s)
Colon/citología , Grasas de la Dieta/farmacología , Ácidos Grasos Insaturados/farmacología , Ácidos Grasos/farmacología , Células Caliciformes/efectos de los fármacos , Alimentación Animal/análisis , Animales , Dieta , Grasas de la Dieta/administración & dosificación , Ácidos Grasos/administración & dosificación , Ácidos Grasos Insaturados/administración & dosificación , Células Caliciformes/metabolismo , Células HT29 , Humanos , Mucina 5AC/genética , Mucina 5AC/metabolismo , Mucina 2/genética , Mucina 2/metabolismo , Aceites de Plantas/administración & dosificación , Aceites de Plantas/química , Ratas , Ratas Wistar
3.
FASEB J ; 27(9): 3860-70, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23756648

RESUMEN

Environmental contaminants are suspected to be involved in the epidemic incidence of metabolic disorders, food ingestion being a primarily route of exposure. We hypothesized that life-long consumption of a high-fat diet that contains low doses of pollutants will aggravate metabolic disorders induced by obesity itself. Mice were challenged from preconception throughout life with a high-fat diet containing pollutants commonly present in food (2,3,7,8-tetrachlorodibenzo-p-dioxin, polychlorinated biphenyl 153, diethylhexyl phthalate, and bisphenol A), added at low doses in the tolerable daily intake range. We measured several blood parameters, glucose and insulin tolerance, hepatic lipid accumulation, and gene expression in adult mice. Pollutant-exposed mice exhibited significant sex-dependent metabolic disorders in the absence of toxicity and weight gain. In males, pollutants increased the expression of hepatic genes (from 36 to 88%) encoding proteins related to cholesterol biosynthesis and decreased (40%) hepatic total cholesterol levels. In females, there was a marked deterioration of glucose tolerance, which may be related to the 2-fold induction of estrogen sulfotransferase and reduced expression of estrogen receptor α (25%) and estrogen target genes (>34%). Because of the very low doses of pollutants used in the mixture, these findings may have strong implications in terms of understanding the potential role of environmental contaminants in food in the development of metabolic diseases.


Asunto(s)
Hígado/efectos de los fármacos , Hígado/metabolismo , Animales , Compuestos de Bencidrilo/toxicidad , Western Blotting , Peso Corporal/efectos de los fármacos , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Fenoles/toxicidad , Bifenilos Policlorados/toxicidad , Dibenzodioxinas Policloradas/toxicidad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
4.
Br J Nutr ; 112(4): 520-35, 2014 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-24932525

RESUMEN

Dairy products derived from the milk of cows fed in pastures are characterised by higher amounts of conjugated linoleic acid and α-linolenic acid (ALA), and several studies have shown their ability to reduce cardiovascular risk. However, their specific metabolic effects compared with standard dairy in a high-fat diet (HFD) context remain largely unknown; this is what we determined in the present study with a focus on the metabolic and intestinal parameters. The experimental animals were fed for 12 weeks a HFD containing 20 % fat in the form of a pasture dairy cream (PDC) or a standard dairy cream (SDC). Samples of plasma, liver, white adipose tissue, duodenum, jejunum and colon were analysed. The PDC mice, despite a higher food intake, exhibited lower fat mass, plasma and hepatic TAG concentrations, and inflammation in the adipose tissue than the SDC mice. Furthermore, they exhibited a higher expression of hepatic PPARα mRNA and adipose tissue uncoupling protein 2 mRNA, suggesting an enhanced oxidative activity of the tissues. These results might be explained, in part, by the higher amounts of ALA in the PDC diet and in the liver and adipose tissue of the PDC mice. Moreover, the PDC diet was found to increase the proportions of two strategic cell populations involved in the protective function of the intestinal epithelium, namely Paneth and goblet cells in the small intestine and colon, compared with the SDC diet. In conclusion, a PDC HFD leads to improved metabolic outcomes and to a stronger gut barrier compared with a SDC HFD. This may be due, at least in part, to the protective mechanisms induced by specific lipids.


Asunto(s)
Bovinos/fisiología , Dieta/veterinaria , Grasas de la Dieta/uso terapéutico , Alimentos Funcionales , Leche , Obesidad/fisiopatología , Paniculitis/prevención & control , Tejido Adiposo Blanco/inmunología , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo Blanco/patología , Crianza de Animales Domésticos , Animales , Productos Lácteos/efectos adversos , Productos Lácteos/análisis , Dieta Alta en Grasa/efectos adversos , Grasas de la Dieta/efectos adversos , Grasas de la Dieta/análisis , Grasas de la Dieta/metabolismo , Femenino , Alimentos Funcionales/análisis , Hipertrigliceridemia/etiología , Hipertrigliceridemia/prevención & control , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Mucosa Intestinal/fisiopatología , Lactancia , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Leche/efectos adversos , Leche/química , Leche/metabolismo , Obesidad/inmunología , Obesidad/metabolismo , Obesidad/patología , Paniculitis/etiología , Componentes Aéreos de las Plantas/química , Componentes Aéreos de las Plantas/crecimiento & desarrollo , Poaceae/química , Poaceae/crecimiento & desarrollo , Distribución Aleatoria
5.
J Lipid Res ; 53(10): 2069-2080, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22865918

RESUMEN

Dietary intake of long-chain n-3 PUFA is now widely advised for public health and in medical practice. However, PUFA are highly prone to oxidation, producing potentially deleterious 4-hydroxy-2-alkenals. Even so, the impact of consuming oxidized n-3 PUFA on metabolic oxidative stress and inflammation is poorly described. We therefore studied such effects and hypothesized the involvement of the intestinal absorption of 4-hydroxy-2-hexenal (4-HHE), an oxidized n-3 PUFA end-product. In vivo, four groups of mice were fed for 8 weeks high-fat diets containing moderately oxidized or unoxidized n-3 PUFA. Other mice were orally administered 4-HHE and euthanized postprandially versus baseline mice. In vitro, human intestinal Caco-2/TC7 cells were incubated with 4-hydroxy-2-alkenals. Oxidized diets increased 4-HHE plasma levels in mice (up to 5-fold, P < 0.01) compared with unoxidized diets. Oxidized diets enhanced plasma inflammatory markers and activation of nuclear factor kappaB (NF-κB) in the small intestine along with decreasing Paneth cell number (up to -19% in the duodenum). Both in vivo and in vitro, intestinal absorption of 4-HHE was associated with formation of 4-HHE-protein adducts and increased expression of glutathione peroxidase 2 (GPx2) and glucose-regulated protein 78 (GRP78). Consumption of oxidized n-3 PUFA results in 4-HHE accumulation in blood after its intestinal absorption and triggers oxidative stress and inflammation in the upper intestine.


Asunto(s)
Aldehídos/farmacocinética , Dieta Alta en Grasa , Ácidos Grasos Omega-3/metabolismo , Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Estrés Oxidativo , Aldehídos/administración & dosificación , Animales , Biomarcadores/metabolismo , Células CACO-2 , Chaperón BiP del Retículo Endoplásmico , Glutatión Peroxidasa/metabolismo , Proteínas de Choque Térmico/metabolismo , Humanos , Absorción Intestinal/fisiología , Peroxidación de Lípido , Masculino , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción
6.
Am J Physiol Endocrinol Metab ; 302(3): E374-86, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22094473

RESUMEN

Low-grade inflammation observed in obesity is a risk factor for cardiovascular disease. Recent studies revealed that this would be linked to gut-derived endotoxemia during fat digestion in high-fat diets, but nothing is known about the effect of lipid composition. The study was designed to test the impact of oil composition of high-fat diets on endotoxin metabolism and inflammation in mice. C57/Bl6 mice were fed for 8 wk with chow or isocaloric isolipidic diets enriched with oils differing in fatty acid composition: milk fat, palm oil, rapeseed oil, or sunflower oil. In vitro, adipocytes (3T3-L1) were stimulated or not with lipopolysaccharide (LPS; endotoxin) and incubated with different fatty acids. In mice, the palm group presented the highest level of IL-6 in plasma (P < 0.01) together with the highest expression in adipose tissue of IL-1ß and of LPS-sensing TLR4 and CD14 (P < 0.05). The higher inflammation in the palm group was correlated with a greater ratio of LPS-binding protein (LBP)/sCD14 in plasma (P < 0.05). The rapeseed group resulted in higher sCD14 than the palm group, which was associated with lower inflammation in both plasma and adipose tissue despite higher plasma endotoxemia. Taken together, our results reveal that the palm oil-based diet resulted in the most active transport of LPS toward tissues via high LBP and low sCD14 and the greatest inflammatory outcomes. In contrast, a rapeseed oil-based diet seemed to result in an endotoxin metabolism driven toward less inflammatory pathways. This shows that dietary fat composition can contribute to modulate the onset of low-grade inflammation through the quality of endotoxin receptors.


Asunto(s)
Tejido Adiposo Blanco/inmunología , Citocinas/metabolismo , Dieta Alta en Grasa/efectos adversos , Enfermedades Metabólicas/etiología , Enfermedades Metabólicas/inmunología , Receptores Inmunológicos/metabolismo , Células 3T3-L1 , Proteínas de Fase Aguda , Tejido Adiposo Blanco/metabolismo , Animales , Biomarcadores/sangre , Biomarcadores/metabolismo , Proteínas Portadoras/sangre , Citocinas/sangre , Ácidos Grasos Monoinsaturados , Ácidos Grasos no Esterificados/efectos adversos , Ácidos Grasos no Esterificados/sangre , Bacterias Gramnegativas/inmunología , Bacterias Gramnegativas/aislamiento & purificación , Bacterias Grampositivas/inmunología , Bacterias Grampositivas/aislamiento & purificación , Intestinos/inmunología , Intestinos/microbiología , Intestinos/patología , Receptores de Lipopolisacáridos/sangre , Receptores de Lipopolisacáridos/metabolismo , Masculino , Glicoproteínas de Membrana/sangre , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/microbiología , Ratones , Ratones Endogámicos C57BL , Aceite de Palma , Aceites de Plantas/efectos adversos , Distribución Aleatoria , Aceite de Brassica napus , Aceite de Girasol , Receptor Toll-Like 4/metabolismo
7.
Am J Physiol Endocrinol Metab ; 298(6): E1161-9, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20233941

RESUMEN

Apelin, an adipocyte-secreted factor upregulated by insulin, is increased in adipose tissue (AT) and plasma with obesity. Apelin was recently identified as a new player in the control of glucose homeostasis. However, the regulation of apelin and APJ (apelin receptor) expression in skeletal muscle in relation to insulin resistance or type 2 diabetes is not known. Thus we studied apelin and APJ expression in AT and muscle in different mice models of obesity and in type 2 diabetic patients. In insulin-resistant high-fat (HF)-fed mice, apelin and APJ expression were increased in AT compared with control. This was not the case in AT of highly insulin-resistant db/db mice. In skeletal muscle, apelin expression was similar in control and HF-fed mice and decreased in db/db mice. APJ expression was decreased in both HF-fed and db/db mice. Control subjects and type 2 diabetic patients were subjected to a hyperinsulinemic-euglycemic clamp, and tissues biopsies were obtained before and at the end of the clamp. There was no significant difference in basal apelin and APJ expression in AT and muscle between control and diabetic patients. However, apelin plasma levels were significantly increased in diabetic patients. During the clamp, hyperinsulinemia increased apelin and APJ expression in AT of control but not in diabetic subjects. In muscle, only APJ mRNA levels were increased in control but also in diabetic patients. Taken together, these data show that apelin and APJ expression in mice and humans is regulated in a tissue-dependent manner and according to the severity of insulin resistance.


Asunto(s)
Tejido Adiposo/fisiología , Proteínas Portadoras/biosíntesis , Diabetes Mellitus Tipo 2/metabolismo , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Músculo Esquelético/fisiología , Receptores Acoplados a Proteínas G/biosíntesis , Adipoquinas , Tejido Adiposo/metabolismo , Adulto , Animales , Apelina , Receptores de Apelina , Proteínas Portadoras/genética , Diabetes Mellitus Tipo 2/genética , Femenino , Regulación de la Expresión Génica , Técnica de Clampeo de la Glucosa , Humanos , Insulina/sangre , Resistencia a la Insulina/fisiología , Péptidos y Proteínas de Señalización Intercelular/genética , Leptina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Músculo Esquelético/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores Acoplados a Proteínas G/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Physiol Genomics ; 34(3): 327-37, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18559965

RESUMEN

In this study we have identified the target genes of sterol regulatory element binding protein (SREBP)-1a and SREBP-1c in primary cultures of human skeletal muscle cells, using adenoviral vectors expressing the mature nuclear form of human SREBP-1a or SREBP-1c combined with oligonucleotide microarrays. Overexpression of SREBP-1a led to significant changes in the expression of 1,315 genes (655 upregulated and 660 downregulated), whereas overexpression of SREBP-1c modified the mRNA level of 514 genes (310 upregulated and 204 downregulated). Gene ontology analysis indicated that in human muscle cells SREBP-1a and -1c are involved in the regulation of a large number of genes that are at the crossroads of different functional pathways, several of which are not directly connected with cholesterol and lipid metabolism. Six hundred fifty-two of all genes identified to be differentially regulated on SREBP overexpression had a sterol regulatory element (SRE) motif in their promoter sequences. Among these, 429 were specifically regulated by SREBP-1a, 69 by SREBP-1c, and 154 by both 1a and 1c. Because both isoforms recognize the same binding motif, we determined whether some of these functional differences could depend on the environment of the SRE motifs in the promoters. Results from promoter analysis showed that different combinations of transcription factor binding sites around the SRE binding motifs may determine regulatory networks of transcription that could explain the superposition of lipid and cholesterol metabolism with various other pathways involved in adaptive responses to stress like hypoxia and heat shock, or involvement in the immune response.


Asunto(s)
Regulación de la Expresión Génica , Redes Reguladoras de Genes , Músculo Esquelético/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Secuencia de Bases , Extractos Celulares , Células Cultivadas , Inmunoprecipitación de Cromatina , Femenino , Glucosa/metabolismo , Glucógeno/biosíntesis , Humanos , Immunoblotting , Masculino , Datos de Secuencia Molecular , Fibras Musculares Esqueléticas/metabolismo , Oxidación-Reducción , Regiones Promotoras Genéticas/genética , Isoformas de Proteínas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Transcripción/metabolismo
9.
Biochem Biophys Res Commun ; 372(4): 924-8, 2008 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-18539141

RESUMEN

Beneficial effects of docosahexaenoic acid (DHA) intake in the prevention of cardiovascular diseases are known, and platelets play a crucial role in cardiovascular complications. However, high doses of DHA may increase lipid peroxidation and induce deleterious effects, notably in platelets. This led us to investigate the effect of DHA on gene expression of some enzymes controlling redox status and prostanoid formation in human megakaryoblastic cells (MEG-01 cell line). MEG-01 cells were incubated in presence of DHA (10 and 100 micromol/L) for 6h. DHA enrichment up-regulated glutathione peroxidase-1 and thromboxane synthase mRNA. DHA increased gene catalase expression and up-regulated PPAR beta/delta and PPAR gamma mRNA in presence of high concentration of DHA. In conclusion, our results support an antioxidant mechanism of DHA. The effects of DHA on cellular redox status could, with others, provide an explanation for the beneficial influence of low consumption of DHA on cardiovascular events.


Asunto(s)
Ácidos Docosahexaenoicos/farmacología , Expresión Génica/efectos de los fármacos , Glutatión Peroxidasa/genética , Prostaglandinas/biosíntesis , Ácido Araquidónico/metabolismo , Línea Celular , Ácidos Grasos/análisis , Humanos , Malondialdehído , Megacariocitos/efectos de los fármacos , Megacariocitos/enzimología , Oxidación-Reducción/efectos de los fármacos , PPAR delta/genética , PPAR-beta/genética , Fosfolípidos/química , Prostaglandina-Endoperóxido Sintasas/genética , Prostaglandinas/genética , Glutatión Peroxidasa GPX1
10.
J Mol Med (Berl) ; 84(9): 747-52, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16897074

RESUMEN

Defective fatty acid oxidation in skeletal muscle is one of the possible causes of insulin resistance. Peroxisome proliferator-activated receptor beta activators are strong inducers of fatty acid oxidation. The aim of this study was to verify whether activation of fatty acid oxidation by PPARbeta agonists in human skeletal muscle cells prepared from type 2 diabetic patients could improve the reduced responses to insulin that characterized this cell model. GW0742 (10 nM) significantly increased fatty acid oxidation and oxidative gene expression in myotubes prepared from both healthy subjects and type 2 diabetic patients. In cells from control subjects, incubation with the agonist for 48 h affected neither insulin-induced rate of glycogen synthesis nor the phosphorylation state of protein kinase B (PKB serine 473). Myotubes from type 2 diabetic patients displayed marked reduction in the effects of insulin on glycogen synthesis and on PKB phosphorylation. However, treatment with PPARbeta agonists did not restore these defects. Therefore, these results indicate that induction of fatty acid oxidation with PPARbeta activators during short-term exposition is not sufficient to correct for insulin resistance in muscle cells from type 2 diabetic patients. This suggests that additional studies are needed to better characterize the link between fatty acid oxidation and insulin sensitivity in humans.


Asunto(s)
Diabetes Mellitus Tipo 2/patología , Insulina/metabolismo , Células Musculares/metabolismo , Células Musculares/patología , PPAR delta/metabolismo , PPAR-beta/metabolismo , Ácido Palmítico/metabolismo , Células Cultivadas , Femenino , Glucógeno/biosíntesis , Humanos , Masculino , Persona de Mediana Edad , Células Musculares/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos , PPAR-beta/agonistas , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Tiazoles/farmacología , Factores de Tiempo
11.
Stem Cells Int ; 2017: 9289213, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28321259

RESUMEN

Adipose-derived stem/stromal cells (ASCs) reside in the stromal vascular fraction (SVF) of adipose tissue (AT) and can be easily isolated. However, extraction of the SVF from lipoaspirate is a critical step in generating ASC, and semiautomated devices have been developed to enhance the efficacy and reproducibility of the outcomes and to decrease manipulation and contamination. In this study, we compared the reference method used in our lab for SVF isolation from lipoaspirate, with three medical devices: GID SVF-1™, Puregraft™, and Stem.pras®. Cell yield and their viability were evaluated as well as their phenotype with flow cytometry. Further on, we determined their proliferative potential using population doublings (PD), PD time (PDT), and clonogenicity assay (CFU-F). Finally, we checked their genetic stability using RT-qPCR for TERT mRNA assay and karyotyping as well as their multilineage potential including adipogenic, chondrogenic, and osteogenic differentiation. Our results demonstrate that all the devices allow the production of SVF cells with consistent yield and viability, in less time than the reference method. Expanded cells from the four methods showed no significant differences in terms of phenotype, proliferation capabilities, differentiation abilities, and genetic stability.

13.
PLoS One ; 9(10): e110653, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25337938

RESUMEN

Inflammation is now recognized as a major factor contributing to type 2 diabetes (T2D). However, while the mechanisms and consequences associated with white adipose tissue inflammation are well described, very little is known concerning the situation in skeletal muscle. The aim of this study was to investigate, in vitro and in vivo, how skeletal muscle inflammation develops and how in turn it modulates local and systemic insulin sensitivity in different mice models of T2D and in humans, focusing on the role of the chemokine MCP1. Here, we found that skeletal muscle inflammation and macrophage markers are increased and associated with insulin resistance in mice models and humans. In addition, we demonstrated that intra-muscular TNFα expression is exclusively restricted to the population of intramuscular leukocytes and that the chemokine MCP1 was associated with skeletal muscle inflammatory markers in these models. Furthermore, we demonstrated that exposure of C2C12 myotubes to palmitate elevated the production of the chemokine MCP1 and that the muscle-specific overexpression of MCP1 in transgenic mice induced the local recruitment of macrophages and altered local insulin sensitivity. Overall our study demonstrates that skeletal muscle inflammation is clearly increased in the context of T2D in each one of the models we investigated, which is likely consecutive to the lipotoxic environment generated by peripheral insulin resistance, further increasing MCP1 expression in muscle. Consequently, our results suggest that MCP1-mediated skeletal muscle macrophages recruitment plays a role in the etiology of T2D.


Asunto(s)
Quimiocina CCL2/fisiología , Resistencia a la Insulina/inmunología , Macrófagos/inmunología , Músculo Esquelético/inmunología , Miositis/inmunología , Animales , Línea Celular , Movimiento Celular , Diabetes Mellitus Tipo 2/inmunología , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Obesos , Ratones Transgénicos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Miositis/metabolismo
14.
PLoS One ; 8(2): e56258, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23437106

RESUMEN

BACKGROUND: Two recent studies demonstrated that bariatric surgery induced remission of type 2 diabetes very soon after surgery and far too early to be attributed to weight loss. In this study, we sought to explore the mechanism/s of this phenomenon by testing the effects of proteins from the duodenum-jejunum conditioned-medium (CM) of db/db or Swiss mice on glucose uptake in vivo in Swiss mice and in vitro in both Swiss mice soleus and L6 cells. We studied the effect of sera and CM proteins from insulin resistant (IR) and insulin-sensitive subjects on insulin signaling in human myoblasts. METHODOLOGY/PRINCIPAL FINDINGS: db/db proteins induced massive IR either in vivo or in vitro, while Swiss proteins did not. In L6 cells, only db/db proteins produced a noticeable increase in basal (473)Ser-Akt phosphorylation, lack of GSK3ß inhibition and a reduced basal (389)Thr-p70-S6K1 phosphorylation. Human IR serum markedly increased basal (473)Ser-Akt phosphorylation in a dose-dependent manner. Human CM IR proteins increased by about twofold both basal and insulin-stimulated (473)Ser-Akt. Basal (9)Ser-GSK3ß phosphorylation was increased by IR subjects serum with a smaller potentiating effect of insulin. CONCLUSIONS: These findings show that jejunal proteins either from db/db mice or from insulin resistant subjects impair muscle insulin signaling, thus inducing insulin resistance.


Asunto(s)
Diabetes Mellitus Experimental/patología , Resistencia a la Insulina , Insulina/metabolismo , Yeyuno/metabolismo , Proteínas/metabolismo , Transducción de Señal , Adulto , Animales , Diferenciación Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Desoxiglucosa/metabolismo , Diabetes Mellitus Experimental/enzimología , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Ratones , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo
15.
Nutr Metab (Lond) ; 10(1): 23, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23413782

RESUMEN

BACKGROUND: Dietary intake of n-3 polyunsaturated fatty acids (PUFA) is primarily recognized to protect against cardiovascular diseases, cognitive dysfunctions and the onset of obesity and associated metabolic disorders. However, some of their properties such as bioavailability can depend on their chemical carriers. The objective of our study was to test the hypothesis that the nature of n-3 PUFA carrier results in different metabolic effects related to adiposity, oxidative stress and inflammation. METHODS: 4 groups of C57BL/6 mice were fed for 8 weeks low fat (LF) diet or high-fat (HF, 20%) diets. Two groups of high-fat diets were supplemented with long-chain n-3 PUFA either incorporated in the form of phospholipids (HF-ω3PL) or triacylglycerols (HF-ω3TG). RESULTS: Both HF-ω3PL and HF-ω3TG diets reduced the plasma concentrations of (i) inflammatory markers such as monocyte chemoattractant protein-1 (MCP-1) and interleukin 6 (IL-6), (ii) leptin and (iii) 4-hydroxy-2-nonenal (4-HNE), a marker of n-6 PUFA-derived oxidative stress compared with the control HF diet. Moreover, in both HF-ω3PL and HF-ω3TG groups, MCP-1 and IL-6 gene expressions were decreased in epididymal adipose tissue and the mRNA level of gastrointestinal glutathione peroxidase GPx2, an antioxidant enzyme, was decreased in the jejunum compared with the control HF diet. The type of n-3 PUFA carrier affected other outcomes. The phospholipid form of n-3 PUFA increased the level of tocopherols in epididymal adipose tissue compared with HF-ω3TG and resulted in smaller adipocytes than the two others HF groups. Adipocytes in the HF-ω3PL and LF groups were similar in size distribution. CONCLUSION: Supplementation of mice diet with long-chain n-3 PUFA during long-term consumption of high-fat diets had the same lowering effects on inflammation regardless of triacyglycerol or phospholipid carrier, whereas the location of these fatty acids on a PL carrier had a major effect on decreasing the size of adipocytes that was not observed with the triacyglycerol carrier. Altogether, these results would support the development functional foods containing LC n-3 PUFA in the form of PL in order to prevent some deleterious outcomes associated with the development of obesity.

16.
J Clin Endocrinol Metab ; 98(2): 802-10, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23284008

RESUMEN

CONTEXT: The hypothesis of a limited expansion of sc adipose tissue during weight gain provides an attractive explanation for the reorientation of excess lipids toward ectopic sites, contributing to visceral adipose depots and metabolic syndrome. OBJECTIVE: Our objective was to define whether the characteristics of sc adipose tissue influence the partition of lipids toward abdominal fat depots during weight gain in healthy men. RESEARCH DESIGN AND METHODS: Forty-one healthy nonobese volunteers performed a 56-day overfeeding protocol (+760 kcal/d). Insulin sensitivity was estimated by euglycemic hyperinsulinemic clamp. Changes in abdominal visceral and sc adipose tissue depots were measured by magnetic resonance imaging. The fate of ingested lipids before and after overfeeding was investigated using a [d31]palmitate test meal, and gene expression was measured by real-time PCR in sc fat biopsies. RESULTS: Overfeeding led to a 2.5-kg body weight increase with large interindividual variations in abdominal sc and visceral adipose tissues. There was no relationship between the relative expansions of these 2 depots, but the increase in visceral depot was positively associated with the magnitude of the postprandial exogenous fatty acid release in the circulation during the test meal. The regulation of lipid storage-related genes (DGAT2, SREBP1c, and CIDEA) was defective in the sc fat of the subjects exhibiting the largest accumulation in visceral depot. CONCLUSIONS: Characteristics of sc adipose tissue appear therefore to contribute to the development of visceral fat depot, supporting the adipose tissue expandability theory and extending it to early stages of weight gain in nonobese subjects.


Asunto(s)
Grasa Intraabdominal/metabolismo , Metabolismo de los Lípidos/fisiología , Hipernutrición/metabolismo , Grasa Subcutánea/metabolismo , Adulto , Expresión Génica , Técnica de Clampeo de la Glucosa , Humanos , Resistencia a la Insulina/fisiología , Masculino , Obesidad/metabolismo
17.
PLoS One ; 8(5): e63997, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23734181

RESUMEN

The intake of ω-3 polyunsaturated fatty acids (PUFAs), which are abundant in marine fish meat and oil, has been shown to exert many beneficial effects. The mechanisms behind those effects are numerous, including interference with the arachidonic acid cascade that produces pro-inflammatory eicosanoids, formation of novel bioactive lipid mediators, and change in the pattern of secreted adipocytokines. In our study, we show that eicosapentaenoic acid (EPA) increases secreted adiponectin from 3T3-L1 adipocytes and in plasma of mice as early as 4 days after initiation of an EPA-rich diet. Using 3T3-L1 adipocytes, we report for the first time that 15-deoxy-δ(12,14)-PGJ3 (15d-PGJ3), a product of EPA, also increases the secretion of adiponectin. We demonstrate that the increased adiponectin secretion induced by 15d-PGJ3 is partially peroxisome proliferator-activated receptor-gamma (PPAR-γ)-mediated. Finally, we show that 3T3-L1 adipocytes can synthesize 15d-PGJ3 from EPA. 15d-PGJ3 was also detected in adipose tissue from EPA-fed mice. Thus, these studies provide a novel mechanism(s) for the therapeutic benefits of ω-3 polyunsaturated fatty acids dietary supplementation.


Asunto(s)
Adipocitos/efectos de los fármacos , Adiponectina/metabolismo , PPAR gamma/metabolismo , Prostaglandina D2/análogos & derivados , Células 3T3-L1 , Adipocitos/metabolismo , Adiponectina/sangre , Adiponectina/genética , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Anilidas/farmacología , Animales , Grasas de la Dieta/administración & dosificación , Ácido Eicosapentaenoico/administración & dosificación , Ácido Eicosapentaenoico/metabolismo , Ácido Eicosapentaenoico/farmacología , Epidídimo/efectos de los fármacos , Epidídimo/metabolismo , Cromatografía de Gases y Espectrometría de Masas , Masculino , Ratones , PPAR gamma/antagonistas & inhibidores , PPAR gamma/genética , Prostaglandina D2/metabolismo , Prostaglandina D2/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Nutr Res ; 33(11): 952-60, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24176235

RESUMEN

Animal studies using a high-fat diet (HFD) have studied the effects of lipid overconsumption by comparing a defined HFD either with a natural-ingredient chow diet or with a defined low-fat diet (LFD), despite the dramatic differences between these control diets. We hypothesized that these differences in the control diet could modify the conclusions regarding the effects that an increase of fat in the diet has on several metabolic parameters. For 11 weeks, C57bl6/J mice were fed a low-fat chow diet (8% energy from fat), a typical semisynthetic LFD (12%), or a semisynthetic HFD (sy-HF) (40%). Conclusions about the effect of sy-HF on body weight gain, subcutaneous adipose tissue, insulin sensitivity, and adipose tissue inflammation were modified according to the control LFD. Conversely, conclusions about epididymal and retroperitoneal adipose tissue; fat intake effects on liver and muscular lipids, cholesterol, free fatty acids, and markers of low-grade inflammation; and of adipose tissue macrophage infiltration were the same regardless of the use of low-fat chow diet or semisynthetic LFD. For some physiological outcomes, conflicting conclusions were even reached about the effects of increased fat intake according to the chosen low-fat control. Some deleterious effects of sy-HF may not be explained by lipid overconsumption but rather by the overall quality of ingredients in a semisynthetic diet. According to the control LFD chosen, conclusions on the lipid-related effects of HFDs must be formulated with great care because some end points are profoundly affected by the ingredient composition of the diet rather than by fat content.


Asunto(s)
Tejido Adiposo/metabolismo , Adiposidad , Investigación Biomédica/métodos , Dieta Alta en Grasa/efectos adversos , Inflamación/etiología , Resistencia a la Insulina , Metabolismo de los Lípidos/efectos de los fármacos , Animales , Dieta con Restricción de Grasas/normas , Grasas de la Dieta/administración & dosificación , Ingestión de Energía , Hígado/efectos de los fármacos , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Grasa Subcutánea/metabolismo , Aumento de Peso
19.
J Clin Endocrinol Metab ; 97(9): 3326-32, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22745235

RESUMEN

CONTEXT: Dietary fibers have been associated with a reduced incidence of type 2 diabetes mellitus in epidemiological studies; however, the precise mechanisms are unknown. OBJECTIVE: The objective of the study was to evaluate the efficacy and site of action of an insoluble dietary fiber derived from maize (HAM-RS2) in improving insulin resistance in subjects at increased risk of type 2 diabetes mellitus. DESIGN: This study was a randomized, controlled crossover, dietary intervention study. SETTING: The study was conducted at the Centre for Diabetes, Endocrinology, and Research, Royal Surrey County Hospital, Guildford, United Kingdom. PARTICIPANTS: Fifteen men and women with insulin resistance participated in the study. INTERVENTION: The intervention included 40 g/d HAM-RS2 compared with a matched placebo for 8 wk. MAIN OUTCOME MEASURES: After each supplement, participants underwent a two-step hyperinsulinemic-euglycemic clamp study with the addition of glucose tracers; a meal tolerance test; arteriovenous sampling across forearm muscle tissue; and a sc adipose tissue biopsy for assessment of gene expression. RESULTS: There was enhanced uptake of glucose into the forearm muscle measured by arteriovenous sampling (65 ± 15% increase after resistant starch; P < 0.001). Adipose tissue function was also affected, with enhanced fatty acid suppression after HAM-RS2 treatment and an increase in gene expression for hormone sensitive lipase (P = 0.005), perilipin (P = 0.011), lipoprotein lipase (P = 0.014), and adipose triglyceride lipase (P = 0.03) in biopsy samples. There was no effect on the insulin sensitivity of hepatic glucose production or plasma lipids after HAM-RS2. CONCLUSION: HAM-RS2 improved peripheral but not hepatic insulin resistance and requires further study as an intervention in patients with or at risk for type 2 diabetes.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Fibras de la Dieta/farmacología , Resistencia a la Insulina/fisiología , Síndrome Metabólico/dietoterapia , Síndrome Metabólico/metabolismo , Músculos/efectos de los fármacos , Músculos/metabolismo , Adulto , Anciano , Biomarcadores/análisis , Biomarcadores/metabolismo , Biopsia , Citocinas/metabolismo , Dieta , Femenino , Expresión Génica/efectos de los fármacos , Glucosa/metabolismo , Técnica de Clampeo de la Glucosa , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/metabolismo , Masculino , Potenciales de la Membrana/efectos de los fármacos , Síndrome Metabólico/fisiopatología , Persona de Mediana Edad , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Método Simple Ciego , Zea mays/química
20.
PLoS One ; 7(7): e40306, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22808131

RESUMEN

Dioxins are persistent organic pollutants interfering with endocrine systems and causing reproductive and developmental disorders. The objective of our project was to determine the impact of an in utero exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on reproductive function of male and female offspring in the rat with a special emphasis on the immature period. We used a low dose of TCDD (unique exposure by oral gavage of 200 ng/kg at 15.5 days of gestation) in order to mirror a response to an environmental dose of TCDD not altering fertility of the progeny. We choose a global gene expression approach using Affymetrix microarray analysis, and testes of 5 days and ovaries of 14 days of age. Less than 1% of the expressed genes in gonads were altered following embryonic TCDD exposure; specifically, 113 genes in ovaries and 56 in testes with 7 genes common to both sex gonads. It included the repressor of the aryl hydrocarbon receptor (Ahrr), the chemokines Ccl5 and Cxcl4 previously shown to be regulated by dioxin in testis, Pgds2/Hpgds and 3 others uncharacterized. To validate and extend the microarray data we realized real-time PCR on gonads at various developmental periods of interest (from 3 to 25 days for ovaries, from 5 to the adult age for testes). Overall, our results evidenced that both sex gonads responded differently to TCDD exposure. For example, we observed induction of the canonic battery of TCDD-induced genes coding enzymes of the detoxifying machinery in ovaries aged of 3-14 days of age (except Cyp1a1 induced at 3-10 days) but not in testes of 5 days (except Ahrr). We also illustrated that inflammatory pathway is one pathway activated by TCDD in gonads. Finally, we identified several new genes targeted by TCDD including Fgf13 in testis and one gene, Ptgds2/Hpgds regulated in the two sex gonads.


Asunto(s)
Embrión de Mamíferos/metabolismo , Perfilación de la Expresión Génica , Ovario/crecimiento & desarrollo , Dibenzodioxinas Policloradas/toxicidad , Efectos Tardíos de la Exposición Prenatal/genética , Caracteres Sexuales , Testículo/crecimiento & desarrollo , Animales , Animales Recién Nacidos , Quimiocinas/genética , Quimiocinas/metabolismo , Cruzamientos Genéticos , Embrión de Mamíferos/efectos de los fármacos , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Ovario/efectos de los fármacos , Ovario/metabolismo , Hipófisis/efectos de los fármacos , Hipófisis/metabolismo , Embarazo , Regiones Promotoras Genéticas/genética , Ratas , Ratas Sprague-Dawley , Reproducción/efectos de los fármacos , Reproducción/genética , Programas Informáticos , Testículo/efectos de los fármacos , Testículo/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA