Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Br J Nutr ; 119(12): 1393-1399, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29845904

RESUMEN

Blueberry, rich in antioxidant and anti-inflammatory phytochemicals, has been demonstrated to lower inflammatory status in adipose induced by high-fat diet (HFD) and obesity. The effect of blueberry on systemic immune functions has not been examined. C57BL/6 mice were randomised to one of three diets - low-fat diet (LFD), HFD and HFD plus 4 % (w/w) blueberry (HFD+B) - for 8 or 12 weeks. Ex vivo T-cell mitogens (concanavalin A (Con A); phytohaemagglutinin), T-cell antibody (anti-CD3; anti-CD3/CD28)-stimulated T-cell proliferation and cytokine production were assessed. After 8 weeks, both HFD groups weighed more (>4 g) than the LFD group; after 12 weeks, HFD+B-fed mice weighed more (>6 g) and had 41 % more adipose tissue than HFD-fed mice (P<0·05). After 12 weeks, T-cell proliferation was less in both HFD groups, compared with the LFD group. HFD-associated decrements in T-cell proliferation were partially (10-50 %) prevented by blueberry supplementation. At 12 weeks, splenocytes from HFD mice, but not from HFD+B mice, produced 51 % less IL-4 (CD3/CD28) and 57 % less interferon-γ (Con A) compared with splenocytes from LFD mice (P<0·05). In response to lipopolysaccharide challenge, splenocytes from both HFD groups produced 24-30 % less IL-6 and 27-33 % less TNF-α compared with splenocytes from LFD mice (P<0·05), indicating impaired acute innate immune response. By demonstrating deleterious impacts of HFD feeding on T-cell proliferation and splenocyte immune responses, our results provide insights into how HFD/obesity can disrupt systemic immune function. The protective effects of blueberry suggest that dietary blueberry can buttress T-cell and systemic immune function against HFD-obesity-associated insults.


Asunto(s)
Arándanos Azules (Planta) , Suplementos Dietéticos , Obesidad/dietoterapia , Obesidad/inmunología , Linfocitos T/inmunología , Adiposidad , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Proliferación Celular , Citocinas/biosíntesis , Dieta con Restricción de Grasas , Dieta Alta en Grasa/efectos adversos , Inmunidad Celular , Inmunosupresores/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Linfocitos T/patología , Aumento de Peso
2.
PLoS One ; 10(8): e0135758, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26284788

RESUMEN

Obesity is a risk factor for colorectal cancer (CRC), and alterations in the colonic microbiome and metabolome may be mechanistically involved in this relationship. The relative contribution of diet and obesity per se are unclear. We compared the effect of diet- and genetically-induced obesity on the intestinal microbiome and metabolome in a mouse model of CRC. Apc1638N mice were made obese by either high fat (HF) feeding or the presence of the Leprdb/db (DbDb) mutation. Intestinal tumors were quantified and stool microbiome and metabolome were profiled. Genetic obesity, and to a lesser extent HF feeding, promoted intestinal tumorigenesis. Each induced distinct microbial patterns: taxa enriched in HF were mostly Firmicutes (6 of 8) while those enriched in DbDb were split between Firmicutes (7 of 12) and Proteobacteria (5 of 12). Parabecteroides distasonis was lower in tumor-bearing mice and its abundance was inversely associated with colonic Il1b production (p<0.05). HF and genetic obesity altered the abundance of 49 and 40 fecal metabolites respectively, with 5 in common. Of these 5, adenosine was also lower in obese and in tumor-bearing mice (p<0.05) and its concentration was inversely associated with colonic Il1b and Tnf production (p<0.05). HF and genetic obesity differentially alter the intestinal microbiome and metabolome. A depletion of adenosine and P.distasonis in tumor-bearing mice could play a mechanistic role in tumor formation. Adenosine and P. distasonis have previously been shown to be anti-inflammatory in the colon and we postulate their reduction could promote tumorigenesis by de-repressing inflammation.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Heces/química , Heces/microbiología , Metaboloma , Microbiota , Obesidad/genética , Receptores de Leptina/genética , Animales , Femenino , Neoplasias Intestinales/genética , Neoplasias Intestinales/metabolismo , Neoplasias Intestinales/microbiología , Masculino , Metaboloma/efectos de los fármacos , Metaboloma/genética , Ratones , Microbiota/efectos de los fármacos , Microbiota/genética , Mutación , Obesidad/etiología , Obesidad/metabolismo , Obesidad/microbiología , Receptores de Leptina/deficiencia
3.
Proc Natl Acad Sci U S A ; 111(14): 5313-8, 2014 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-24706839

RESUMEN

The HLA-F adjacent transcript 10 (FAT10) is a member of the ubiquitin-like gene family that alters protein function/stability through covalent ligation. Although FAT10 is induced by inflammatory mediators and implicated in immunity, the physiological functions of FAT10 are poorly defined. We report the discovery that FAT10 regulates lifespan through pleiotropic actions on metabolism and inflammation. Median and overall lifespan are increased 20% in FAT10ko mice, coincident with elevated metabolic rate, preferential use of fat as fuel, and dramatically reduced adiposity. This phenotype is associated with metabolic reprogramming of skeletal muscle (i.e., increased AMP kinase activity, ß-oxidation and -uncoupling, and decreased triglyceride content). Moreover, knockout mice have reduced circulating glucose and insulin levels and enhanced insulin sensitivity in metabolic tissues, consistent with elevated IL-10 in skeletal muscle and serum. These observations suggest novel roles of FAT10 in immune metabolic regulation that impact aging and chronic disease.


Asunto(s)
Adiposidad/genética , Longevidad/genética , Ubiquitinas/genética , Adipocitos/metabolismo , Animales , Biomarcadores/metabolismo , Metabolismo Energético , Femenino , Masculino , Ratones , Ratones Noqueados , Oxidación-Reducción , Triglicéridos/metabolismo
4.
Obesity (Silver Spring) ; 22(6): 1485-94, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24616441

RESUMEN

OBJECTIVE: To investigate the role of TNF-like weak inducer of apoptosis (TWEAK) in pathological adipose tissue (AT) remodeling and complications of obesity. METHODS: Wild type (WT) and TWEAK knockout (KO) mice were fed normal diet (ND) or a high fat diet (HFD) for up to 17 weeks. Adipocyte death was induced using an established transgenic mouse model of inducible adipocyte apoptosis (FAT-ATTAC). Metabolic, biochemical, histologic, and flow cytometric analyses were performed. RESULTS: TWEAK and its receptor, fibroblast growth factor-inducible molecule 14 (Fn14) were upregulated in gonadal (g)AT of WT mice after HFD week 4 and 24 h after induction of adipocyte apoptosis. Phenotypes of KO and WT mouse were indistinguishable through HFD week 8. However, at week 17 obese KO mice had ∼30% larger gAT adipocytes and gAT mass than WT mice, coincident with reduced adipocyte death, enhanced insulin signaling, Th2/M2 immune skewing, fewer thick collagen fibers, and altered expression of extracellular matrix constituents and modulators that is consistent with reduced fibrosis and larger adipocytes. KO mice were less steatotic and became more insulin sensitive and glucose tolerant than WT mice after HFD week 12. CONCLUSION: TWEAK constrains "healthy" gAT expansion and promotes metabolic complications in severe obesity.


Asunto(s)
Tejido Adiposo/metabolismo , Apoptosis/fisiología , Eliminación de Gen , Obesidad Mórbida/genética , Obesidad Mórbida/prevención & control , Factores de Necrosis Tumoral/genética , Adipocitos/metabolismo , Animales , Citocina TWEAK , Dieta Alta en Grasa , Resistencia a la Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal , Receptor de TWEAK , Factores de Necrosis Tumoral/metabolismo , Regulación hacia Arriba
5.
Lipids ; 49(5): 415-22, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24595513

RESUMEN

Individuals with type 2 diabetes mellitus (T2DM) are at increased risk of developing cardiovascular disease (CVD), possibly associated with elevated plasma free fatty acid concentrations. Paradoxically, evidence suggests that unsaturated, compared to saturated fatty acids, suppress macrophage cholesterol efflux, favoring cholesterol accumulation in the artery wall. Murine bone marrow-derived macrophages (BMDM) were used to further explore the relationship between saturated and unsaturated fatty acids, and cholesterol efflux mediated by ATP-binding cassette transporters (ABCA1 and ABCG1) through transcription factors liver-x-receptor-alpha (LXR-α) and sterol receptor element binding protein (SREBP)-1. BMDM isolated from C57BL/6 mice were exposed to 100 µM linoleic acid (18:2) or palmitic acid (16:0) for 16 h, and 25 µg/mL oxidized low density lipoprotein for an additional 24 h. ABCA1 and ABCG1 mRNA expression was suppressed to a greater extent by 18:2 (60 % and 54 %, respectively) than 16:0 (30 % and 29 %, respectively) relative to the control (all p < 0.01). 18:2 decreased ABCA1 protein levels by 94 % and high density lipoprotein (HDL) mediated cholesterol efflux by 53 % (both p < 0.05), and had no significant effect on ABCG1, LXR-α or SREBP-1 protein levels. 16:0 had no effect on ABCA1, ABCG1, LXR-α or SREBP-1 protein expression or HDL-mediated cholesterol efflux. These results suggest that 18:2, relative to 16:0, attenuated macrophage HDL-mediated cholesterol efflux through down regulation of ABCA1 mRNA and protein levels but not through changes in LXR-α or SREBP-1 expression. The effect of 18:2 relative to 16:0 on macrophages cholesterol homeostasis may exacerbate the predisposition of individuals with T2DM to increased CVD risk.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Células de la Médula Ósea/citología , Colesterol/metabolismo , Ácido Linoleico/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Relación Estructura-Actividad
6.
Cancer Prev Res (Phila) ; 6(12): 1304-16, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24085778

RESUMEN

Obesity is associated with increased risk in hepatocellular carcinoma (HCC) development and mortality. An important disease control strategy is the prevention of obesity-related hepatic inflammation and tumorigenesis by dietary means. Here, we report that apo-10'-lycopenoic acid (APO10LA), a cleavage metabolite of lycopene at its 9',10'-double bond by carotene-9',10'-oxygenase, functions as an effective chemopreventative agent against hepatic tumorigenesis and inflammation. APO10LA treatment on human liver THLE-2 and HuH7 cells dose dependently inhibited cell growth and upregulated sirtuin 1 (SIRT1), a NAD(+)-dependent protein deacetylase that may suppress hepatic carcinogenesis. This observed SIRT1 induction was associated with decreased cyclin D1 protein, increased cyclin-dependent kinase inhibitor p21 protein expression, and induced apoptosis. APO10LA supplementation (10 mg/kg diet) for 24 weeks significantly reduced diethylnitrosamine-initiated, high fat diet (HFD)-promoted hepatic tumorigenesis (50% reduction in tumor multiplicity; 65% in volume) and lung tumor incidence (85% reduction) in C57Bl/6J mice. The chemopreventative effects of APO10LA were associated with increased hepatic SIRT1 protein and deacetylation of SIRT1 targets, as well as with decreased caspase-1 activation and SIRT1 protein cleavage. APO10LA supplementation in diet improved glucose intolerance and reduced hepatic inflammation [decreased inflammatory foci, TNFα, interleukin (IL)-6, NF-κB p65 protein expression, and STAT3 activation] in HFD-fed mice. Furthermore, APO10LA suppressed Akt activation, cyclin D1 gene, and protein expression and promoted PARP protein cleavage in transformed cells within liver tumors. Taken together, these data indicate that APO10LA can effectively inhibit HFD-promoted hepatic tumorigenesis by stimulating SIRT1 signaling while reducing hepatic inflammation.


Asunto(s)
Carcinoma Hepatocelular/prevención & control , Carotenoides/uso terapéutico , Transformación Celular Neoplásica/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Dietilnitrosamina/toxicidad , Ácidos Grasos Insaturados/uso terapéutico , Inflamación/prevención & control , Neoplasias Hepáticas/prevención & control , Alquilantes/toxicidad , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Carcinoma Hepatocelular/inducido químicamente , Carcinoma Hepatocelular/patología , Carotenoides/metabolismo , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/inducido químicamente , Transformación Celular Neoplásica/patología , Ciclina D1/genética , Ciclina D1/metabolismo , Femenino , Prueba de Tolerancia a la Glucosa , Humanos , Inflamación/inducido químicamente , Inflamación/patología , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/patología , Licopeno , Ratones , Ratones Endogámicos C57BL , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Proc Natl Acad Sci U S A ; 110(13): 5133-8, 2013 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-23479618

RESUMEN

Patients with type 2 diabetes (T2D) have disease-associated changes in B-cell function, but the role these changes play in disease pathogenesis is not well established. Data herein show B cells from obese mice produce a proinflammatory cytokine profile compared with B cells from lean mice. Complementary in vivo studies show that obese B cell-null mice have decreased systemic inflammation, inflammatory B- and T-cell cytokines, adipose tissue inflammation, and insulin resistance (IR) compared with obese WT mice. Reduced inflammation in obese/insulin resistant B cell-null mice associates with an increased percentage of anti-inflammatory regulatory T cells (Tregs). This increase contrasts with the sharply decreased percentage of Tregs in obese compared with lean WT mice and suggests that B cells may be critical regulators of T-cell functions previously shown to play important roles in IR. We demonstrate that B cells from T2D (but not non-T2D) subjects support proinflammatory T-cell function in obesity/T2D through contact-dependent mechanisms. In contrast, human monocytes increase proinflammatory T-cell cytokines in both T2D and non-T2D analyses. These data support the conclusion that B cells are critical regulators of inflammation in T2D due to their direct ability to promote proinflammatory T-cell function and secrete a proinflammatory cytokine profile. Thus, B cells are potential therapeutic targets for T2D.


Asunto(s)
Linfocitos B/inmunología , Citocinas/inmunología , Diabetes Mellitus Tipo 2/inmunología , Obesidad/inmunología , Linfocitos T Reguladores/inmunología , Animales , Linfocitos B/patología , Diabetes Mellitus Tipo 2/patología , Diabetes Mellitus Tipo 2/terapia , Femenino , Humanos , Inflamación/inmunología , Inflamación/patología , Inflamación/terapia , Masculino , Ratones , Ratones Obesos , Obesidad/patología , Obesidad/terapia , Linfocitos T Reguladores/patología
9.
Mol Aspects Med ; 34(1): 59-70, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23068072

RESUMEN

When humans eat more and exercise less, they tend to become obese and unhealthy. The molecular pathways that link obesity to serious diseases like Type 2 diabetes and cardiovascular disease have become a subject of intensive scientific investigation because the exploding prevalence of obesity worldwide represents a grave new threat to the health of hundreds of millions of people. However, obesity is not always destiny. Two important clinical populations have been valuable to understand the mechanisms behind this conundrum: individuals who exhibit metabolic dysfunction, diabetes and elevated cardiovascular disease risk despite a lean body type, and individuals who are relatively protected from these dangers despite significant obesity. Study of this second group of 'metabolically healthy obese' people in particular has been revealing because such individuals exhibit specific, identifiable, anatomic, cellular and molecular features that set them apart from the rest of us who suffer declining health with increasing weight. Here, we examine some of these features, including some mouse models that are informative of mechanism, and suggest hypotheses for further study, including the possibility that genes and pathways of the immune system might offer new diagnostic or therapeutic targets.


Asunto(s)
Obesidad/metabolismo , Adipocitos/citología , Adipocitos/metabolismo , Tejido Adiposo/química , Animales , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/fisiopatología , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/fisiopatología , Ejercicio Físico , Humanos , Inflamación/etiología , Inflamación/fisiopatología , Resistencia a la Insulina , Síndrome Metabólico/etiología , Síndrome Metabólico/fisiopatología , Ratones , Obesidad/complicaciones
10.
Mol Aspects Med ; 34(1): 39-58, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23159341

RESUMEN

The human gut is densely populated by commensal and symbiotic microbes (the "gut microbiota"), with the majority of the constituent microorganisms being bacteria. Accumulating evidence indicates that the gut microbiota plays a significant role in the development of obesity, obesity-associated inflammation and insulin resistance. In this review we discuss molecular and cell biological mechanisms by which the microbiota participate in host functions that impact the development and maintenance of the obese state, including host ingestive behavior, energy harvest, energy expenditure and fat storage. We additionally explore the diverse signaling pathways that regulate gut permeability and bacterial translocation to the host and how these are altered in the obese state to promote the systemic inflammation ("metabolic endotoxemia") that is a hallmark of obesity and its complications. Fundamental to our discussions is the concept of "crosstalk", i.e., the biochemical exchange between host and microbiota that maintains the metabolic health of the superorganism and whose dysregulation is a hallmark of the obese state. Differences in community composition, functional genes and metabolic activities of the gut microbiota appear to distinguish lean vs obese individuals, suggesting that gut 'dysbiosis' contributes to the development of obesity and/or its complications. The current challenge is to determine the relative importance of obesity-associated compositional and functional changes in the microbiota and to identify the relevant taxa and functional gene modules that promote leanness and metabolic health. As diet appears to play a predominant role in shaping the microbiota and promoting obesity-associated dysbiosis, parallel initiatives are required to elucidate dietary patterns and diet components (e.g., prebiotics, probiotics) that promote healthy gut microbiota. How the microbiota promotes human health and disease is a rich area of investigation that is likely to generate fundamental discoveries in energy metabolism, molecular endocrinology and immunobiology and may lead to new strategies for prevention of obesity and its complications.


Asunto(s)
Tracto Gastrointestinal/microbiología , Resistencia a la Insulina , Metagenoma , Obesidad/metabolismo , Animales , Endotoxemia/microbiología , Endotoxemia/fisiopatología , Ingestión de Energía , Metabolismo Energético , Tracto Gastrointestinal/fisiopatología , Humanos , Inflamación/microbiología , Inflamación/fisiopatología , Ratones , Obesidad/microbiología , Obesidad/fisiopatología , Prebióticos/análisis , Probióticos/administración & dosificación , Delgadez/metabolismo
11.
Endocrinology ; 153(9): 4266-77, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22778213

RESUMEN

Menopause promotes central obesity, adipose tissue (AT) inflammation, and insulin resistance (IR). Both obesity and the loss of estrogen can activate innate and adaptive immune cells (macrophages, T cells). The respective impacts of weight gain and loss of ovarian hormones on AT inflammation and IR are poorly understood. Here we determined the temporal kinetics of fat accretion, AT inflammation, and IR over a 26-wk time course in ovariectomized (OVX) mice, a model of menopause. OVX and sham-operated (SHM) C57BL6 mice were fed a normal chow diet. Weight, body composition (magnetic resonance imaging), total and regional adiposity, activity, food intake, AT crown-like structures, biohumoral measures, and insulin sensitivity (insulin tolerance testing and homeostatic model assessment) were determined at wk 12, 20, and 26. Macrophages and T cells from perigonadal AT were immunophenotyped by fluorescence-associated cell sorting, and perigonadal adipose tissue (PGAT) gene expression was quantified by quantitative PCR. OVX mice (≈ 31 g) became fatter than SHM mice (≈ 26 g) by wk 12, but mice were equally insulin sensitive. PGAT of OVX mice contained more T cells but expressed higher levels of M2-MΦ (arginase-1) and T cell-regulatory (cytotoxic T-lymphocyte antigen 4) genes. At wk 20, both OVX and SHM mice weighed approximately 35 g and were equally insulin sensitive with comparable amounts of PGAT and total body fat. OVX mice became less insulin sensitive than SHM mice by wk 26, coincident with the down-regulation of PGAT arginase-1 (-20-fold) and cytotoxic T-lymphocyte antigen 4 (2-fold) and up-regulation of M1/Th1 genes CD11c (+2-fold), IL12p40 (+2-fold), and interferon-γ (+78-fold). Ovarian hormone loss in mice induces PGAT inflammation and IR by mechanisms that can be uncoupled from OVX-induced obesity.


Asunto(s)
Tejido Adiposo/inmunología , Tejido Adiposo/metabolismo , Adiposidad/fisiología , Resistencia a la Insulina/inmunología , Ovariectomía , Animales , Composición Corporal/fisiología , Peso Corporal/fisiología , Femenino , Citometría de Flujo , Inmunohistoquímica , Ratones , Reacción en Cadena de la Polimerasa
12.
EMBO Rep ; 13(2): 150-6, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22222206

RESUMEN

Sqstm1/p62 functions in the non-canonical activation of nuclear factor (erythroid-derived 2)-like 2 (Nrf2). However, its physiological relevance is not certain. Here, we show that p62(-/-) mice exhibited an accelerated presentation of ageing phenotypes, and tissues from these mice created a pro-oxidative environment owing to compromised mitochondrial electron transport. Accordingly, mitochondrial function rapidly declined with age in p62(-/-) mice. In addition, p62 enhanced basal Nrf2 activity, conferring a higher steady-state expression of NAD(P)H dehydrogenase, quinone 1 (Nqo1) to maintain mitochondrial membrane potential and, thereby, restrict excess oxidant generation. Together, the p62-Nrf2-Nqo1 cascade functions to assure mammalian longevity by stabilizing mitochondrial integrity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas del Citoesqueleto/metabolismo , Proteínas de Choque Térmico/metabolismo , Longevidad/fisiología , Mamíferos/fisiología , Mitocondrias/metabolismo , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Animales , Autofagia , Femenino , Proteínas de Choque Térmico/deficiencia , Proteína 1 Asociada A ECH Tipo Kelch , Masculino , Ratones , Oxidación-Reducción , Proteína Sequestosoma-1 , Transducción de Señal
13.
J Clin Invest ; 121(6): 2102-10, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21633178

RESUMEN

Lipid droplets (LDs) are intracellular organelles that store neutral lipids within cells. Over the last two decades there has been a dramatic growth in our understanding of LD biology and, in parallel, our understanding of the role of LDs in health and disease. In its simplest form, the LD regulates the storage and hydrolysis of neutral lipids, including triacylglycerol and/or cholesterol esters. It is becoming increasingly evident that alterations in the regulation of LD physiology and metabolism influence the risk of developing metabolic diseases such as diabetes. In this review we provide an update on the role of LD-associated proteins and LDs in metabolic disease.


Asunto(s)
Tejido Adiposo/fisiopatología , Lípidos/fisiología , Enfermedades Metabólicas/fisiopatología , Vacuolas/fisiología , Adipocitos/fisiología , Adipocitos/ultraestructura , Animales , Diabetes Mellitus/fisiopatología , Metabolismo Energético/fisiología , Ácidos Grasos/efectos adversos , Ácidos Grasos/metabolismo , Hígado Graso/fisiopatología , Humanos , Resistencia a la Insulina , Péptidos y Proteínas de Señalización Intracelular/fisiología , Lipólisis/fisiología , Ratones , Ratones Mutantes , Músculo Esquelético/metabolismo , Enfermedad del Hígado Graso no Alcohólico , Obesidad/fisiopatología , Transducción de Señal/fisiología , Triglicéridos/metabolismo
14.
Diabetes ; 60(4): 1168-76, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21346175

RESUMEN

OBJECTIVE: Obesity-associated low-grade systemic inflammation resulting from increased adipose mass is strongly related to the development of insulin resistance and type 2 diabetes as well as other metabolic complications. Recent studies have demonstrated that the obese metabolic state can be improved by ablating certain inflammatory signaling pathways. Tumor progression locus 2 (TPL2), a kinase that integrates signals from Toll receptors, cytokine receptors, and inhibitor of κ-B kinase-ß is an important regulator of inflammatory pathways. We used TPL2 knockout (KO) mice to investigate the role of TPL2 in mediating obesity-associated inflammation and insulin resistance. RESEARCH DESIGN AND METHODS: Male TPL2KO and wild-type (WT) littermates were fed a low-fat diet or a high-fat diet to investigate the effect of TPL2 deletion on obesity, inflammation, and insulin sensitivity. RESULTS: We demonstrate that TPL2 deletion does not alter body weight gain or adipose depot weight. However, hyperinsulinemic euglycemic clamp studies revealed improved insulin sensitivity with enhanced glucose uptake in skeletal muscle and increased suppression of hepatic glucose output in obese TPL2KO mice compared with obese WT mice. Consistent with an improved metabolic phenotype, immune cell infiltration and inflammation was attenuated in the adipose tissue of obese TPL2KO mice coincident with reduced hepatic inflammatory gene expression and lipid accumulation. CONCLUSIONS: Our results provide the first in vivo demonstration that TPL2 ablation attenuates obesity-associated metabolic dysfunction. These data suggest TPL2 is a novel target for improving the metabolic state associated with obesity.


Asunto(s)
Resistencia a la Insulina/fisiología , Quinasas Quinasa Quinasa PAM/metabolismo , Obesidad/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Tejido Adiposo/inmunología , Tejido Adiposo/metabolismo , Animales , Western Blotting , Células de la Médula Ósea , Células Cultivadas , Grasas de la Dieta/efectos adversos , Hígado Graso/genética , Hígado Graso/metabolismo , Femenino , Humanos , Inmunohistoquímica , Inflamación/genética , Inflamación/fisiopatología , Resistencia a la Insulina/genética , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Hígado/metabolismo , Hígado/patología , Quinasas Quinasa Quinasa PAM/genética , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Noqueados , Obesidad/inducido químicamente , Obesidad/genética , Proteínas Proto-Oncogénicas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Triglicéridos/metabolismo
15.
Diabetes ; 59(5): 1171-81, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20185806

RESUMEN

OBJECTIVE: To identify, localize, and determine M1/M2 polarization of epidydimal adipose tissue (eAT) macrophages (Phis) during high-fat diet (HFD)-induced obesity. RESEARCH DESIGN AND METHODS: Male C57BL/6 mice were fed an HFD (60% fat kcal) or low-fat diet (LFD) (10% fat kcal) for 8 or 12 weeks. eATMPhis (F4/80(+) cells) were characterized by in vivo fluorescent labeling, immunohistochemistry, fluorescence-activated cell sorting, and quantitative PCR. RESULTS: Recruited interstitial macrophage galactose-type C-type lectin (MGL)1(+)/CD11c(-) and crown-like structure-associated MGL1(-)/CD11c(+) and MGL1(med)/CD11c(+) eATMPhis were identified after 8 weeks of HFD. MGL1(med)/CD11c(+) cells comprised approximately 65% of CD11c(+) eATMPhis. CD11c(+) eATMPhis expressed a mixed M1/M2 profile, with some M1 transcripts upregulated (IL-12p40 and IL-1beta), others downregulated (iNOS, caspase-1, MCP-1, and CD86), and multiple M2 and matrix remodeling transcripts upregulated (arginase-1, IL-1Ra, MMP-12, ADAM8, VEGF, and Clec-7a). At HFD week 12, each eATMPhi subtype displayed an enhanced M2 phenotype as compared with HFD week 8. CD11c(+) subtypes downregulated IL-1beta and genes mediating antigen presentation (I-a, CD80) and upregulated the M2 hallmark Ym-1 and genes promoting oxidative metabolism (PGC-1alpha) and adipogenesis (MMP-2). MGL1(med)/CD11c(+) eATMPhis upregulated additional M2 genes (IL-13, SPHK1, CD163, LYVE-1, and PPAR-alpha). MGL1(med)/CD11c(+) ATMPhis expressing elevated PGC-1alpha, PPAR-alpha, and Ym-1 transcripts were selectively enriched in eAT of obese mice fed pioglitazone for 6 days, confirming the M2 features of the MGL1(med)/CD11c(+) eATMPhi transcriptional profile and implicating PPAR activation in its elicitation. CONCLUSIONS: These results 1) redefine the phenotypic potential of CD11c(+) eATMPhis and 2) suggest previously unappreciated phenotypic and functional commonality between murine and human ATMPhis in the development of obesity and its complications.


Asunto(s)
Tejido Adiposo/metabolismo , Antígeno CD11c/metabolismo , Grasas de la Dieta/efectos adversos , Macrófagos/metabolismo , Obesidad/inducido químicamente , Obesidad/metabolismo , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Antígeno B7-2/metabolismo , Citometría de Flujo , Inmunohistoquímica , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Subunidad p40 de la Interleucina-12/metabolismo , Interleucina-13/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/fisiopatología , Reacción en Cadena de la Polimerasa , Receptores de Superficie Celular/metabolismo
16.
Obesity (Silver Spring) ; 18(10): 1918-25, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20111012

RESUMEN

The role of adaptive immunity in obesity-associated adipose tissue (AT) inflammation and insulin resistance (IR) is controversial. We employed flow cytometry and quantitative PCR to assess T-cell recruitment and activation in epididymal AT (eAT) of C57BL/6 mice during 4-22 weeks of a high-fat diet (HFD (60% energy)). By week 6, eAT mass and stromal vascular cell (SVC) number increased threefold in mice fed HFD, coincident with onset of IR. We observed no increase in the proportion of CD3(+) SVCs or in gene expression of CD3, interferon-γ (IFN-γ), or regulated upon activation, normal T-cell expressed and secreted (RANTES) during the first 16 weeks of HFD. In contrast, CD11c(+) macrophages (MΦ) were enriched sixfold by week 8 (P < 0.01). SVC enrichment for T cells (predominantly CD4(+) and CD8(+)) and elevated IFN-γ and RANTES gene expression were detected by 20-22 weeks of HFD (P < 0.01), coincident with the resolution of eAT remodeling. HFD-induced T-cell priming earlier in the obesity time course is suggested by (i) elevated (fivefold) interleukin-12 (IL-12)p40 gene expression in eAT by week 12 (P ≤ 0.01) and (ii) greater IFN-γ secretion from phorbol myristate acetate (PMA)/ionophore-stimulated eAT explants at week 6 (onefold, P = 0.08) and week 12 (fivefold, P < 0.001). In conclusion, T-cell enrichment and IFN-γ gene induction occur subsequent to AT macrophage (ATMΦ) recruitment, onset of IR and resolution of eAT remodeling. However, enhanced priming for IFN-γ production suggests the contribution of CD4(+) and/or CD8(+) effectors to cell-mediated immune responses promoting HFD-induced AT inflammation and IR.


Asunto(s)
Tejido Adiposo/inmunología , Inflamación/etiología , Resistencia a la Insulina , Macrófagos/metabolismo , Obesidad/inmunología , Linfocitos T/metabolismo , Células TH1/metabolismo , Tejido Adiposo/metabolismo , Animales , Antígeno CD11c/metabolismo , Complejo CD3/metabolismo , Linfocitos T CD8-positivos/metabolismo , Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Grasas de la Dieta/efectos adversos , Expresión Génica , Inflamación/inmunología , Interferón gamma/genética , Interferón gamma/metabolismo , Subunidad p40 de la Interleucina-12/genética , Subunidad p40 de la Interleucina-12/metabolismo , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Obesidad/metabolismo , Acetato de Tetradecanoilforbol/farmacología
17.
J Lipid Res ; 51(5): 975-82, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-19797618

RESUMEN

Perilipin A is the most abundant phosphoprotein on adipocyte lipid droplets and is essential for lipid storage and lipolysis. Perilipin null mice exhibit diminished adipose tissue, elevated basal lipolysis, reduced catecholamine-stimulated lipolysis, and increased insulin resistance. To understand the physiological consequences of increased perilipin expression in vivo, we generated transgenic mice that overexpressed either human or mouse perilipin using the adipocyte-specific aP2 promoter/enhancer. Phenotypes of female transgenic and wild-type mice were characterized on chow and high-fat diets (HFDs). When challenged with an HFD, transgenic mice exhibited lower body weight, fat mass, and adipocyte size than wild-type mice. Expression of oxidative genes was increased and lipogenic genes decreased in brown adipose tissue of transgenic mice. Basal and catecholamine-stimulated lipolysis was decreased and glucose tolerance significantly improved in transgenic mice fed a HFD. Perilipin overexpression in adipose tissue protects against HFD-induced adipocyte hypertrophy, obesity, and glucose intolerance. Alterations in brown adipose tissue metabolism may mediate the effects of perilipin overexpression on body fat, although the mechanisms by which perilipin overexpression alters brown adipose tissue metabolism remain to be determined. Our findings demonstrate a novel role for perilipin expression in adipose tissue metabolism and regulation of obesity and its metabolic complications.


Asunto(s)
Dieta/efectos adversos , Obesidad/genética , Obesidad/prevención & control , Fosfoproteínas/genética , Adipocitos/metabolismo , Adipocitos/patología , Animales , Proteínas Portadoras , Catecolaminas/farmacología , Tamaño de la Célula , Grasas de la Dieta/efectos adversos , Femenino , Expresión Génica , Glucosa/metabolismo , Homeostasis/genética , Humanos , Insulina/metabolismo , Lipólisis/efectos de los fármacos , Lipólisis/genética , Masculino , Ratones , Ratones Transgénicos , Obesidad/etiología , Obesidad/metabolismo , Especificidad de Órganos , Oxidación-Reducción , Perilipina-1 , Aumento de Peso/efectos de los fármacos , Aumento de Peso/genética
18.
Biochem Biophys Res Commun ; 392(1): 1-3, 2010 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-19850007

RESUMEN

Menopause, the age-related loss of ovarian hormone production, promotes increased adiposity and associated metabolic pathology, but molecular mechanisms remain unclear. We previously reported that estrogen increases skeletal muscle PPARdelta expression in vivo, and transgenic mice overexpressing muscle-specific PPARdelta are reportedly protected from diet-induced obesity. We thus hypothesized that obesity observed in ovariectomized mice, a model of menopause, may result in part from abrogated expression of muscle PPARdelta and/or downstream mediators such as FoxO1. To test this hypothesis, we ovariectomized (OVX) or sham-ovariectomized (SHM) 10-week old female C57Bl/6J mice, and subsequently harvested quadriceps muscles 12weeks later for gene expression studies. Compared to SHM, muscle from OVX mice displayed significantly decreased expression of PPARdelta (3.4-fold), FoxO1 (4.5-fold), PDK-4 (2.3-fold), and UCP-2 (1.8-fold). Consistent with studies indicating PPARdelta and FoxO1 regulate muscle fiber type, we observed dramatic OVX-specific decreases in slow isoforms of the contractile proteins myosin light chain (11.1-fold) and troponin C (11.8-fold). In addition, muscles from OVX mice expressed 57% less myogenin (drives type I fiber formation), 2-fold more MyoD (drives type II fiber formation), and 1.6-fold less musclin (produced exclusively by type II fibers) than SHM, collectively suggesting a shift towards less type I oxidative fibers. Finally, and consistent with changes in PPARdelta and FoxO1 activity, we observed decreased expression of atrogin-1 (2.3-fold) and MuRF-1 (1.9-fold) in OVX mice. In conclusion, muscles from ovariectomized mice display decreased PPARdelta and FoxO1 expression, abrogated expression of downstream targets involved in lipid and protein metabolism, and gene expression profiles indicating less type I oxidative fibers.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Menopausia/metabolismo , Músculo Esquelético/metabolismo , Obesidad/genética , Ovario/metabolismo , PPAR delta/metabolismo , Animales , Femenino , Proteína Forkhead Box O1 , Metabolismo de los Lípidos/genética , Menopausia/genética , Ratones , Ratones Endogámicos C57BL , Fibras Musculares de Contracción Lenta/metabolismo , Proteína MioD/genética , Miogenina/genética
19.
J Nutr ; 139(8): 1510-6, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19515743

RESUMEN

Adipose tissue (AT) inflammation promotes insulin resistance (IR) and other obesity complications. AT inflammation and IR are associated with oxidative stress, adipocyte death, and the scavenging of dead adipocytes by proinflammatory CD11c+ AT macrophages (ATMPhi). We tested the hypothesis that supplementation of an obesitogenic (high-fat) diet with whole blueberry (BB) powder protects against AT inflammation and IR. Male C57Bl/6j mice were maintained for 8 wk on 1 of 3 diets: low-fat (10% of energy) diet (LFD), high-fat (60% of energy) diet (HFD) or the HFD containing 4% (wt:wt) whole BB powder (1:1 Vaccinium ashei and V. corymbosum) (HFD+B). BB supplementation (2.7% of total energy) did not affect HFD-associated alterations in energy intake, metabolic rate, body weight, or adiposity. We observed an emerging pattern of gene expression in AT of HFD mice indicating a shift toward global upregulation of inflammatory genes (tumor necrosis factor-alpha, interleukin-6, monocyte chemoattractant protein 1, inducible nitric oxide synthase), increased M1-polarized ATMPhi (CD11c+), and increased oxidative stress (reduced glutathione peroxidase 3). This shift was attenuated or nonexistent in HFD+B-fed mice. Furthermore, mice fed the HFD+B were protected from IR and hyperglycemia coincident with reductions in adipocyte death. Salutary effects of BB on adipocyte physiology and ATMPhi gene expression may reflect the ability of BB anthocyanins to alter mitogen-activated protein kinase and nuclear factor-kappaB stress signaling pathways, which regulate cell fate and inflammatory genes. These results suggest that cytoprotective and antiinflammatory actions of dietary BB can provide metabolic benefits to combat obesity-associated pathology.


Asunto(s)
Adipocitos/efectos de los fármacos , Antocianinas/farmacología , Antiinflamatorios/farmacología , Arándanos Azules (Planta) , Muerte Celular/efectos de los fármacos , Resistencia a la Insulina , Preparaciones de Plantas/farmacología , Adiposidad/efectos de los fármacos , Animales , Arándanos Azules (Planta)/química , Muerte Celular/genética , Quimiocina CCL2/metabolismo , Dieta , Grasas de la Dieta/administración & dosificación , Frutas , Expresión Génica/efectos de los fármacos , Glutatión Peroxidasa/metabolismo , Hiperglucemia/prevención & control , Interleucina-6/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo II/metabolismo , Preparaciones de Plantas/química , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
20.
Endocrinology ; 150(5): 2161-8, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19179442

RESUMEN

Menopause, an age-related loss of ovarian hormone production, promotes increased adiposity and insulin resistance. However, the diet-independent mechanism by which loss of ovarian function promotes increased adipose tissue mass and associated metabolic pathologies remains unclear. To address this question, we monitored food intake and weight gain of ovariectomized (OVX) mice and sham OVX (SHM) mice for 12 wk. Although food intake was similar, OVX mice gained 25% more weight than SHM mice. Moreover, the OVX mice accumulated 4.7- and 4.4-fold more perigonadal and inguinal adipose tissue by weight, respectively, with 4.4-fold (perigonadal, P < 0.001) and 5.3-fold (inguinal, P < 0.01) larger adipocytes and no change in adipocyte cell number. OVX-induced adiposity was coincident with an 18% decrease in metabolic rate during the dark phase (P = 0.001) as well as an 11% decrease during the light phase (P = 0.03). In addition, ambulatory activity levels of OVX mice were decreased only during the dark phase (40%, P = 0.008). OVX mice displayed evidence of immune infiltration and inflammation in adipose tissue, because perigonadal and inguinal adipose depots from OVX mice had increased expression of TNFalpha, iNOS, CD11c, and other hallmarks of adipose tissue inflammation. In contrast, expression of the T cell marker CD3 (3.5-fold, P = 0.03) and Th1 cytokine interferon-gamma (IFNgamma) (2.6-fold, P = 0.02) were elevated in perigonadal but not sc fat. Finally, histology revealed OVX-specific liver hepatic steatosis, coincident with increased PPARgamma gene expression and downstream lipogenic gene expression. In summary, OVX in mice decreases energy expenditure, without altering energy intake, resulting in adipocyte hypertrophy, adipose tissue inflammation, and hepatic steatosis.


Asunto(s)
Metabolismo Energético , Inflamación/etiología , Obesidad/etiología , Ovariectomía/efectos adversos , Tejido Adiposo/patología , Animales , Distribución de la Grasa Corporal , Peso Corporal/fisiología , Ingestión de Energía/fisiología , Metabolismo Energético/fisiología , Ayuno/sangre , Ayuno/metabolismo , Femenino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/fisiología , Obesidad/inmunología , Obesidad/metabolismo , Obesidad/patología , Tamaño de los Órganos , Factores de Tiempo , Útero/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...