Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Int J Obes (Lond) ; 48(7): 964-972, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38459259

RESUMEN

BACKGROUND/OBJECTIVES: Proinflammatory cytokines are increased in obese adipose tissue, including inflammasome key masters. Conversely, IL-18 protects against obesity and metabolic dysfunction. We focused on the IL-18 effect in controlling adipose tissue remodeling and metabolism. MATERIALS/SUBJECTS AND METHODS: We used C57BL/6 wild-type (WT) and interleukine-18 deficient (IL-18-/-) male mice fed a chow diet and samples from bariatric surgery patients. RESULTS: IL-18-/- mice showed increased adiposity and proinflammatory cytokine levels in adipose tissue, leading to glucose intolerance. IL-18 was widely secreted by stromal vascular fraction but not adipocytes from mice's fatty tissue. Chimeric model experiments indicated that IL-18 controls adipose tissue expansion through its presence in tissues other than bone marrow. However, IL-18 maintains glucose homeostasis when present in bone marrow cells. In humans with obesity, IL-18 expression in omental tissue was not correlated with BMI or body fat mass but negatively correlated with IRS1, GLUT-4, adiponectin, and PPARy expression. Also, the IL-18RAP receptor was negatively correlated with IL-18 expression. CONCLUSIONS: IL-18 signaling may control adipose tissue expansion and glucose metabolism, as its absence leads to spontaneous obesity and glucose intolerance in mice. We suggest that resistance to IL-18 signaling may be linked with worse glucose metabolism in humans with obesity.


Asunto(s)
Tejido Adiposo , Interleucina-18 , Ratones Endogámicos C57BL , Obesidad , Animales , Interleucina-18/metabolismo , Ratones , Masculino , Tejido Adiposo/metabolismo , Humanos , Obesidad/metabolismo , Intolerancia a la Glucosa/metabolismo , Modelos Animales de Enfermedad , Ratones Noqueados
2.
Biochim Biophys Acta Mol Basis Dis ; 1863(1): 139-151, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27677832

RESUMEN

Macrophage infiltration into adipose tissue (AT) is a hallmark of the chronic inflammatory response in obesity and is supported by an intense monocyte migration towards AT. Although it has been detected an increased proportion of circulating CD16+ monocyte subsets in obese subjects, the mechanisms underlying this effect and the contribution of these cells to the inflamed profile of obese AT are still poorly understood. We investigated whether factors secreted by human obese omental AT could polarize monocytes to CD16+ enriched phenotype, and how these changes could modify their migratory capacity towards adipose tissue itself. We show that explants of human obese omental AT, obtained during bariatric surgery, released higher levels of MIP1-α, TNFα, leptin and also VEGF, together with increasing amounts of microparticles (MP), when compared to explants of lean subcutaneous AT. A higher content of circulating MP derived from preadipocytes and leukocytes was also detected in plasma of obese subjects. Conditioned media or MP released from obese omental AT increased CD16 and CCR5 expression on CD14+CD16- monocytes and augmented their migratory capacity towards the conditioned media from obese omental AT, itself. This effect was inhibited when MIP1-α was neutralized. Additionally, we demonstrate that MP derived from obese omental AT carry and transfer TLR8 to monocytes, thus triggering an increase in CD16 expression in those cells. Our data shows a positive feedback loop between blood monocytes and obese omental AT, which releases chemotactic mediators and TLR8-enriched MP, thus inducing an up-regulation of CD16+ monocytes, favoring leukocyte infiltration in the obese omental AT.


Asunto(s)
Tejido Adiposo/inmunología , Micropartículas Derivadas de Células/inmunología , Monocitos/inmunología , Obesidad/inmunología , Receptores CCR5/inmunología , Receptores de IgG/inmunología , Receptor Toll-Like 8/inmunología , Tejido Adiposo/patología , Adulto , Micropartículas Derivadas de Células/patología , Femenino , Proteínas Ligadas a GPI/análisis , Proteínas Ligadas a GPI/inmunología , Humanos , Inflamación/inmunología , Inflamación/patología , Masculino , Persona de Mediana Edad , Monocitos/patología , Obesidad/patología , Receptores CCR5/análisis , Receptores de IgG/análisis , Receptor Toll-Like 8/análisis
3.
Nutrition ; 58: 167-174, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30502680

RESUMEN

OBJECTIVE: Chia seed oil is the richest source of plant-based ω-3 fatty acid, α-linolenic acid, but its potential and mechanisms of action to treat obesity are unclear. The aim of the study was to evaluate the effects of chia oil (ChOi) supplementation on body composition and insulin signaling in skeletal muscles of obese mice. METHODS: Male C57 BL/6 mice (n = 8/group) were fed regular control chow or a high-fat diet (HFD) for 135 d. Another HFD group additionally received ChOi from 90 to 135 d. RESULTS: Consumption of ChOi reduced fat mass accumulation and increased lean mass as evidenced by nuclear magnetic resonance. Moreover, obese mice treated with ChOi showed higher tyrosine phosphorylation of insulin receptor substrate 1, greater activation of protein kinase B, and increased translocation of glucose transporter type 4 in skeletal muscle tissue in response to insulin. ChOi supplementation improved glucose levels and insulin tolerance; decreased serum insulin, leptin, and triacylglycerols; and increased blood high-density lipoprotein cholesterol levels. All these effects caused by the use of ChOi seemed to be independent of the resolution of inflammation because the markers of inflammation were not altered in animals fed the HFD. CONCLUSION: The molecular effects observed in muscle tissue together with changes in body composition may have contributed to the increased glucose tolerance and to the healthy phenotype presented by obese animals treated with ChOi.


Asunto(s)
Composición Corporal/efectos de los fármacos , Suplementos Dietéticos , Insulina/metabolismo , Músculo Esquelético/metabolismo , Obesidad/metabolismo , Aceites de Plantas/farmacología , Salvia , Animales , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Músculo Esquelético/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
4.
J Control Release ; 281: 11-18, 2018 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-29753960

RESUMEN

Nanoparticles have specific features (lipophilicity, surface charge, composition and size). Studies regarding the biological behavior of nanoparticles in diseases such diabetics and obesity are scarce. Here, we evaluated two nanoparticles: magnetic core mesoporous silica (MSN) (58 nm) and polycaprolactone (PCL) nanoparticle (280 nm) in obese mice. Changes in the biodistribution were observed, especially considering the mononuclear phagocyte system (MPS), and the visceral fat tissue. Nonetheless, our data corroborates the influence of size in the biodistribution in obese animals, supporting that smaller nanoparticles, may show a higher tissue deposition at spleen, due the associated splenomegaly and the complications arising from this state. Finally, our study demonstrated that, in obesity, probably due the low-grade inflammatory state associated with metabolic syndrome a difference in accumulation of nanoparticles was found, with profound impact in the tissue deposition of nanoparticles.


Asunto(s)
Nanopartículas de Magnetita/química , Obesidad/metabolismo , Poliésteres/química , Dióxido de Silicio/química , Animales , Grasa Intraabdominal/metabolismo , Imagen por Resonancia Magnética/métodos , Magnetismo , Masculino , Ratones Endogámicos C57BL , Sistema Mononuclear Fagocítico/metabolismo , Porosidad , Distribución Tisular
5.
Obesity (Silver Spring) ; 24(12): 2522-2532, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27753270

RESUMEN

OBJECTIVE: To investigate the role of obesity on the bone marrow microenvironment and evaluate its possible impact on the adipogenic potential of mesenchymal stem cells (MSC). METHODS: C57BL/6 male mice were fed with a high-fat diet (HFD) for 10 weeks. Femurs and tibiae were collected, and bone marrow mesenchymal stem cells (BM-MSC) were isolated and analyzed for proliferative potential, immunophenotype, and expression of adipogenesis markers. Their capacity to produce extracellular matrix proteins and proinflammatory cytokines in vitro was also evaluated. RESULTS: HFD mice presented a significant increase in bone marrow cellularity and higher tumor necrosis factor-α production in vitro. BM-MSC from HFD mice had higher proliferative capacity, produced more extracellular matrix proteins associated with adipogenesis, collagen I, and collagen IV, and showed increased constitutive expression of adipogenic markers, peroxisome proliferator-activated receptor-γ, and CCAAT/enhanced binding protein family-α, without changes in preadipocyte factor-1 expression. Incubation with adipocyte-differentiation medium induced further increase in CCAAT/enhanced binding protein family-α and augmented adiponectin expression in obese BM-MSC. These alterations did not result in increased adipogenic differentiation within the bone marrow. Moreover, BM-HSC from HFD mice, co-cultivated with BM-MSCs from lean mice, exerted paracrine effects on these cells, inducing augment of peroxisome proliferator-activated receptor-γ. CONCLUSIONS: The data suggest that obesity promotes an inflammatory microenvironment in bone marrow that commits BM-MSC to adipogenesis.


Asunto(s)
Adipogénesis/fisiología , Células de la Médula Ósea/fisiología , Células Madre Mesenquimatosas/fisiología , Obesidad/fisiopatología , Animales , Médula Ósea , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Diferenciación Celular , Proliferación Celular , Dieta Alta en Grasa , Inflamación , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , PPAR gamma/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis
6.
PLoS One ; 8(5): e64220, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23675529

RESUMEN

We investigated the consequences of mild maternal malnutrition in rat dams, in terms of thymocyte responses and the putative role of leptin. The young progeny of dams submitted to protein deprivation (PD) during lactation showed at 30 days of age lower body and thymus weights, significant alterations in CD4/CD8-defined T cell subsets without modifications in total thymocyte number as well as in proliferative response. Despite, the rats from PD group did not present alterations in leptin circulating levels, the expression of leptin receptor ObRb was enhanced in their thymocytes. This change was accompanied by an increase in leptin signaling response of thymocytes from PD rats, with an increase in JAK2 and STAT3 phosphorylation after leptin stimulation. Thymocytes from PD rats also presented a decreased rate of spontaneous apoptosis when compared to controls. Accordingly, higher expression of anti-apoptotic protein Bcl-2, and lower of pro-apoptotic protein Bax, with no change of pro-apoptotic Bad, and higher pro-caspase 3 content were detected in PD thymocytes. Moreover, thymocytes from PD group exhibited a constitutive higher nuclear content of p65 NF-kB associated to a lower IkB content in the cytoplasm. Finally, although there was no change in ob gene expression in PD thymocytes, a higher mRNA expression for the Ob gene was observed in the thymic microenvironment from PD animals. Taken together, the results show that mild maternal protein deprivation during lactation affects thymic homeostasis, enhancing leptin activity, which in turn protects thymocytes from apoptosis in the young progeny, with possible consequences upon the immune response of these animals in adult life.


Asunto(s)
Apoptosis , Lactancia , Leptina/metabolismo , Desnutrición Proteico-Calórica , Timocitos/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Peso Corporal , Microambiente Celular/genética , Microambiente Celular/inmunología , Dieta con Restricción de Proteínas , Femenino , Regulación de la Expresión Génica , Inmunofenotipificación , Janus Quinasa 2/metabolismo , Leptina/sangre , Leptina/genética , Masculino , FN-kappa B/metabolismo , Ratas , Receptores de Leptina/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Subgrupos de Linfocitos T/metabolismo , Timocitos/inmunología , Timo/citología
7.
Biol Trace Elem Res ; 149(1): 64-70, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22528770

RESUMEN

Iron bioavailability seems to be regulated by vitamin A (VA) but the molecular events involved in this mechanism are not well understood. It is also known that retinoids mediate most of their function via interaction with retinoid receptors, which act as ligand-activated transcription factors controlling the expression of a number of target genes. Here, we evaluated the VA effects on the modulation of the levels of mRNA encoding proteins involved in the iron bioavailability, whether in the intestinal absorption process or in the liver iron metabolism. The expression of genes involved in iron intestinal absorption (divalent metal transporter 1, duodenal cytochrome B, ferroportin 1 FPN1, and ferritin) were evaluated in vitro by treating Caco-2 cells with retinoic acid or in vivo by observing the effects of vitamin A deficiency (VAD) in BALB/C mice. Liver hepcidin and ferritin mRNA levels were upregulated by VAD; however, this condition did not promote any change on the expression of those genes that participate in the iron absorption. Moreover, data from the in vitro analysis showed that VA induced FPN1 gene expression by a hepcidin-independent manner. Therefore, the in vivo results support the idea that VAD may not affect iron absorption but would rather affect iron mobilization mechanisms. On the other hand, our results using Caco-2 cells raises the possibility that VA addition to intestinal epithelium may improve iron absorption through the induction of FPN1 gene expression.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Regulación de la Expresión Génica , Mucosa Intestinal/metabolismo , Hierro/metabolismo , Vitamina A/análogos & derivados , Animales , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/metabolismo , Western Blotting , Células CACO-2 , Proteínas de Transporte de Catión/genética , Suplementos Dietéticos , Diterpenos , Evaluación Preclínica de Medicamentos , Duodeno/efectos de los fármacos , Duodeno/metabolismo , Ferritinas/genética , Ferritinas/metabolismo , Hepcidinas , Humanos , Absorción Intestinal , Mucosa Intestinal/citología , Mucosa Intestinal/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ésteres de Retinilo , Tretinoina/farmacología , Vitamina A/administración & dosificación , Vitamina A/farmacología , Deficiencia de Vitamina A/genética , Deficiencia de Vitamina A/metabolismo
8.
Am J Physiol Endocrinol Metab ; 295(3): E626-36, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18559980

RESUMEN

Epidemiological and experimental studies have demonstrated that early postnatal nutrition has been associated with long-term effects on glucose homeostasis in adulthood. Recently, our group demonstrated that undernutrition during early lactation affects the expression and activation of key proteins of the insulin signaling cascade in rat skeletal muscle during postnatal development. To elucidate the molecular mechanisms by which undernutrition during early life leads to changes in insulin sensitivity in peripheral tissues, we investigated the insulin signaling in adipose tissue. Adipocytes were isolated from epididymal fat pads of adult male rats that were the offspring of dams fed either a normal or a protein-free diet during the first 10 days of lactation. The cells were incubated with 100 nM insulin before the assays for immunoblotting analysis, 2-deoxyglucose uptake, immunocytochemistry for GLUT4, and/or actin filaments. Following insulin stimulation, adipocytes isolated from undernourished rats presented reduced tyrosine phosphorylation of IR and IRS-1 and increased basal phosphorylation of IRS-2, Akt, and mTOR compared with controls. Basal glucose uptake was increased in adipocytes from the undernourished group, and the treatment with LY294002 induced only a partial inhibition both in basal and in insulin-stimulated glucose uptake, suggesting an involvement of phosphoinositide 3-kinase activity. These alterations were accompanied by higher GLUT4 content in the plasma membrane and alterations in the actin cytoskeleton dynamics. These data suggest that early postnatal undernutrition impairs insulin sensitivity in adulthood by promoting changes in critical steps of insulin signaling in adipose tissue, which may contribute to permanent changes in glucose homeostasis.


Asunto(s)
Adipocitos/metabolismo , Dieta con Restricción de Proteínas , Transportador de Glucosa de Tipo 4/metabolismo , Lactancia/fisiología , Proteína Oncogénica v-akt/metabolismo , Proteínas Quinasas/metabolismo , Adipocitos/fisiología , Tejido Adiposo/metabolismo , Animales , Antibióticos Antineoplásicos/farmacología , Western Blotting , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Separación Celular , Cromonas/farmacología , Inhibidores Enzimáticos/farmacología , Femenino , Glucosa/metabolismo , Hipoglucemiantes/farmacología , Inmunohistoquímica , Insulina/metabolismo , Insulina/farmacología , Morfolinas/farmacología , Fosforilación , Transporte de Proteínas , Ratas , Ratas Wistar , Sirolimus/farmacología , Serina-Treonina Quinasas TOR , Tiazolidinas/farmacología , Tirosina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA