Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Hepatol Commun ; 2(3): 313-328, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29507905

RESUMEN

Increasingly, evidence suggests that exposure to maternal obesity creates an inflammatory environment in utero, exerting long-lasting postnatal signatures on the juvenile innate immune system and microbiome that may predispose offspring to development of fatty liver disease. We found that exposure to a maternal Western-style diet (WD) accelerated fibrogenesis in the liver of offspring and was associated with early recruitment of proinflammatory macrophages at 8-12 weeks and microbial dysbiosis as early as 3 weeks of age. We further demonstrated that bone marrow-derived macrophages (BMDMs) were polarized toward an inflammatory state at 8 weeks of age and that a potent antioxidant, pyrroloquinoline quinone (PQQ), reversed BMDM metabolic reprogramming from glycolytic toward oxidative metabolism by restoring trichloroacetic acid cycle function at isocitrate dehydrogenase. This resulted in reduced inflammation and inhibited collagen fibril formation in the liver at 20 weeks of age, even when PQQ was withdrawn at 3 weeks of age. Beginning at 3 weeks of age, WD-fed mice developed a decreased abundance of Parabacteroides and Lactobacillus, together with increased Ruminococcus and decreased tight junction gene expression by 20 weeks, whereas microbiota of mice exposed to PQQ retained compositional stability with age, which was associated with improved liver health. Conclusion: Exposure to a maternal WD induces early gut dysbiosis and disrupts intestinal tight junctions, resulting in BMDM polarization and induction of proinflammatory and profibrotic programs in the offspring that persist into adulthood. Disrupted macrophage and microbiota function can be attenuated by short-term maternal treatment with PQQ prior to weaning, suggesting that reshaping the early gut microbiota in combination with reprogramming macrophages during early weaning may alleviate the sustained proinflammatory environment, preventing the rapid progression of nonalcoholic fatty liver disease to nonalcoholic steatohepatitis in offspring of obese mothers. (Hepatology Communications 2018;2:313-328).

2.
Diabetes ; 67(4): 651-661, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29138256

RESUMEN

Adipose tissue expansion progresses rapidly during postnatal life, influenced by both prenatal maternal factors and postnatal developmental cues. The ratio of omega-6 (n-6) relative to n-3 polyunsaturated fatty acids (PUFAs) is believed to regulate perinatal adipogenesis, but the cellular mechanisms and long-term effects are not well understood. We lowered the fetal and postnatal n-6/n-3 PUFA ratio exposure in wild-type offspring under standard maternal dietary fat amounts to test the effects of low n-6/n-3 ratios on offspring adipogenesis and adipogenic potential. Relative to wild-type pups receiving high perinatal n-6/n-3 ratios, subcutaneous adipose tissue in 14-day-old wild-type pups receiving low n-6/n-3 ratios had more adipocytes that were smaller in size; decreased Pparγ2, Fabp4, and Plin1; several lipid metabolism mRNAs; coincident hypermethylation of the PPARγ2 proximal promoter; and elevated circulating adiponectin. As adults, offspring that received low perinatal n-6/n-3 ratios were diet-induced obesity (DIO) resistant and had a lower positive energy balance and energy intake, greater lipid fuel preference and non-resting energy expenditure, one-half the body fat, and better glucose clearance. Together, the findings support a model in which low early-life n-6/n-3 ratios remodel adipose morphology to increase circulating adiponectin, resulting in a persistent adult phenotype with improved metabolic flexibility that prevents DIO.


Asunto(s)
Adipogénesis , Glucemia/metabolismo , Ácidos Grasos Omega-3/sangre , Ácidos Grasos Omega-6/sangre , Metabolismo de los Lípidos , Obesidad/epidemiología , Efectos Tardíos de la Exposición Prenatal/epidemiología , Adipocitos/citología , Adiponectina/metabolismo , Animales , Animales Recién Nacidos , Proliferación Celular , Tamaño de la Célula , Metilación de ADN , Dieta Alta en Grasa , Grasas de la Dieta , Ingestión de Energía , Metabolismo Energético , Proteínas de Unión a Ácidos Grasos/metabolismo , Femenino , Ratones , Obesidad/sangre , PPAR gamma/metabolismo , Perilipina-1/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/sangre , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Factores de Riesgo
3.
Sci Rep ; 7(1): 18095, 2017 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-29273781

RESUMEN

The intrauterine period is a critical time wherein developmental exposure can influence risk for chronic disease including childhood obesity. Using umbilical cord-derived mesenchymal stem cells (uMSC) from offspring born to normal-weight and obese mothers, we tested the hypothesis that changes in infant body composition over the first 5 months of life correspond with differences in cellular metabolism and transcriptomic profiles at birth. Higher long-chain acylcarnitine concentrations, lipid transport gene expression, and indicators of oxidative stress in uMSC-adipocytes were related to higher adiposity at 5 months of age. In uMSC-myocytes, lower amino acid concentrations and global differential gene expression for myocyte growth, amino acid biosynthesis, and oxidative stress were related to lower infant percent fat-free mass at 5 months of age, particularly in offspring of obese mothers. This is the first evidence of human infant adipocyte- or myocyte-related alterations in cellular metabolic pathways that correspond with increased adiposity and lower fat-free mass in early infancy. These pathways might reflect the effects of an adverse maternal metabolic environment on the fetal metabolome and genome. Our findings suggest that programmed differences in infant stem cell metabolism correspond with differences in body composition in early life, a known contributor to obesity risk.


Asunto(s)
Adiposidad/fisiología , Peso al Nacer/fisiología , Expresión Génica , Células Madre Mesenquimatosas/metabolismo , Índice de Masa Corporal , Diferenciación Celular , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Lactante , Masculino , Metabolómica , Estrés Oxidativo/fisiología
4.
JCI Insight ; 2(21)2017 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-29093265

RESUMEN

Maternal obesity is a global health problem that increases offspring obesity risk. The metabolic pathways underlying early developmental programming in human infants at risk for obesity remain poorly understood, largely due to barriers in fetal/infant tissue sampling. Utilizing umbilical cord-derived mesenchymal stem cells (uMSC) from offspring of normal weight and obese mothers, we tested whether energy metabolism and gene expression differ in differentiating uMSC myocytes and adipocytes, in relation to maternal obesity exposures and/or neonatal adiposity. Biomarkers of incomplete ß-oxidation were uniquely positively correlated with infant adiposity and maternal lipid levels in uMSC myocytes from offspring of obese mothers only. Metabolic and biosynthetic processes were enriched in differential gene expression analysis related to maternal obesity. In uMSC adipocytes, maternal obesity and lipids were associated with downregulation in multiple insulin-dependent energy-sensing pathways including PI3K and AMPK. Maternal lipids correlated with uMSC adipocyte upregulation of the mitochondrial respiratory chain but downregulation of mitochondrial biogenesis. Overall, our data revealed cell-specific alterations in metabolism and gene expression that correlated with maternal obesity and adiposity of their offspring, suggesting tissue-specific metabolic and regulatory changes in these newborn cells. We provide important insight into potential developmental programming mechanisms of increased obesity risk in offspring of obese mothers.


Asunto(s)
Adiposidad/fisiología , Regulación del Desarrollo de la Expresión Génica , Células Madre Mesenquimatosas/metabolismo , Obesidad/complicaciones , Obesidad/metabolismo , Adiposidad/genética , Aminoácidos/análisis , Biomarcadores , Carnitina/análogos & derivados , Carnitina/análisis , Colorado , Transporte de Electrón/genética , Metabolismo Energético/genética , Ácidos Grasos/metabolismo , Femenino , Humanos , Lactante , Recién Nacido , Insulina/metabolismo , Metabolismo de los Lípidos , Lípidos/análisis , Estudios Longitudinales , Masculino , Fenómenos Fisiologicos Nutricionales Maternos , Células Madre Mesenquimatosas/patología , Mitocondrias/genética , Mitocondrias/metabolismo , Enfermedades Mitocondriales , Células Musculares/metabolismo , Células Musculares/patología , Embarazo
5.
FASEB J ; 31(4): 1434-1448, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28007783

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is widespread in adults and children. Early exposure to maternal obesity or Western-style diet (WD) increases steatosis and oxidative stress in fetal liver and is associated with lifetime disease risk in the offspring. Pyrroloquinoline quinone (PQQ) is a natural antioxidant found in soil, enriched in human breast milk, and essential for development in mammals. We investigated whether a supplemental dose of PQQ, provided prenatally in a mouse model of diet-induced obesity during pregnancy, could protect obese offspring from progression of NAFLD. PQQ treatment given pre- and postnatally in WD-fed offspring had no effect on weight gain but increased metabolic flexibility while reducing body fat and liver lipids, compared with untreated obese offspring. Indices of NAFLD, including hepatic ceramide levels, oxidative stress, and expression of proinflammatory genes (Nos2, Nlrp3, Il6, and Ptgs2), were decreased in WD PQQ-fed mice, concomitant with increased expression of fatty acid oxidation genes and decreased Pparg expression. Notably, these changes persisted even after PQQ withdrawal at weaning. Our results suggest that supplementation with PQQ, particularly during pregnancy and lactation, protects offspring from WD-induced developmental programming of hepatic lipotoxicity and may help slow the advancing epidemic of NAFLD in the next generation.-Jonscher, K. R., Stewart, M. S., Alfonso-Garcia, A., DeFelice, B. C., Wang, X. X., Luo, Y., Levi, M., Heerwagen, M. J. R., Janssen, R. C., de la Houssaye, B. A., Wiitala, E., Florey, G., Jonscher, R. L., Potma, E. O., Fiehn, O. Friedman, J. E. Early PQQ supplementation has persistent long-term protective effects on developmental programming of hepatic lipotoxicity and inflammation in obese mice.


Asunto(s)
Antioxidantes/uso terapéutico , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Obesidad/complicaciones , Cofactor PQQ/uso terapéutico , Efectos Tardíos de la Exposición Prenatal/prevención & control , Animales , Antioxidantes/administración & dosificación , Antioxidantes/farmacología , Ceramidas/metabolismo , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Dieta Alta en Grasa/efectos adversos , Suplementos Dietéticos , Femenino , Interleucina-6/genética , Interleucina-6/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/etiología , Obesidad/tratamiento farmacológico , Obesidad/etiología , Estrés Oxidativo , PPAR gamma/metabolismo , Cofactor PQQ/administración & dosificación , Cofactor PQQ/farmacología , Embarazo , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico , Efectos Tardíos de la Exposición Prenatal/etiología
6.
Am J Clin Nutr ; 103(5): 1291-300, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27140533

RESUMEN

BACKGROUND: Increased maternal body mass index (BMI) is a robust risk factor for later pediatric obesity. Accumulating evidence suggests that human milk (HM) may attenuate the transfer of obesity from mother to offspring, potentially through its effects on early development of the infant microbiome. OBJECTIVES: Our objective was to identify early differences in intestinal microbiota in a cohort of breastfeeding infants born to obese compared with normal-weight (NW) mothers. We also investigated relations between HM hormones (leptin and insulin) and both the taxonomic and functional potentials of the infant microbiome. DESIGN: Clinical data and infant stool and fasting HM samples were collected from 18 NW [prepregnancy BMI (in kg/m(2)) <24.0] and 12 obese (prepregnancy BMI >30.0) mothers and their exclusively breastfed infants at 2 wk postpartum. Infant body composition at 2 wk was determined by air-displacement plethysmography. Infant gastrointestinal microbes were estimated by using 16S amplicon and whole-genome sequencing. HM insulin and leptin were determined by ELISA; short-chain fatty acids (SCFAs) were measured in stool samples by using gas chromatography. Power was set at 80%. RESULTS: Infants born to obese mothers were exposed to 2-fold higher HM insulin and leptin concentrations (P < 0.01) and showed a significant reduction in the early pioneering bacteria Gammaproteobacteria (P = 0.03) and exhibited a trend for elevated total SCFA content (P < 0.06). Independent of maternal prepregnancy BMI, HM insulin was positively associated with both microbial taxonomic diversity (P = 0.03) and Gammaproteobacteria (e.g., Enterobacteriaceae; P = 0.04) and was negatively associated with Lactobacillales (e.g., Streptococcaceae; P = 0.05). Metagenomic analysis showed that HM leptin and insulin were associated with decreased bacterial proteases, which are implicated in intestinal permeability, and reduced concentrations of pyruvate kinase, a biomarker of pediatric gastrointestinal inflammation. CONCLUSION: Our results indicate that, although maternal obesity may adversely affect the early infant intestinal microbiome, HM insulin and leptin are independently associated with beneficial microbial metabolic pathways predicted to increase intestinal barrier function and reduce intestinal inflammation. This trial was registered at clinicaltrials.gov as NCT01693406.


Asunto(s)
Microbioma Gastrointestinal , Insulina/análisis , Leptina/análisis , Leche Humana/química , Adulto , Biomarcadores/sangre , Composición Corporal , Índice de Masa Corporal , Lactancia Materna , Estudios de Cohortes , Estudios Transversales , Ácidos Grasos Volátiles/análisis , Heces/química , Femenino , Gammaproteobacteria/aislamiento & purificación , Humanos , Lactante , Lactobacillales/aislamiento & purificación , Modelos Lineales , Masculino , Análisis Multivariante , Obesidad/sangre , Obesidad/prevención & control , Pletismografía , Piruvato Quinasa/sangre , Factores de Riesgo
7.
Atherosclerosis ; 250: 172-9, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27072340

RESUMEN

BACKGROUND AND OBJECTIVE: Atherosclerosis is both a chronic inflammatory disease and a lipid metabolism disorder. C/EBPß is well documented for its role in the development of hematopoietic cells and integration of lipid metabolism. However, C/EBPß's role in atherosclerotic progression has not been examined. We assessed the impact of hematopoietic CEBPß deletion in ApoE(-/-) mice on hyperlipidemia, inflammatory responses and lesion formation in the aorta. METHODS AND RESULTS: ApoE(-/-) mice were reconstituted with bone marrow cells derived from either WT or C/EBPß(-/-) mice and placed on low fat or high fat/high cholesterol diet for 11 weeks. Hematopoietic C/EBPß deletion in ApoE(-/-) mice reduced blood and hepatic lipids and gene expression of hepatic stearoyl CoA desaturase 1 and fatty acid synthase while expression of ATP binding cassette transporter G1, cholesterol 7-alpha-hydroxylase, and liver X receptor alpha genes were significantly increased. ApoE(-/-) mice reconstituted with C/EBPß(-/-) bone marrow cells also significantly reduced blood cytokine levels and reduced lesion area in aortic sinuses compared with ApoE(-/-) mice reconstituted with WT bone marrow cells. Silencing of C/EBPß in RAW264.7 macrophage cells prevented oxLDL-mediated foam cell formation and inflammatory cytokine secretion in conditioned medium. CONCLUSION: C/EBPß in hematopoietic cells is crucial to regulate diet-induced inflammation, hyperlipidemia and atherosclerosis development.


Asunto(s)
Aterosclerosis/metabolismo , Médula Ósea/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Colesterol/sangre , Dieta/efectos adversos , Inflamación/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Medios de Cultivo Condicionados/química , Citocinas/metabolismo , Femenino , Células Espumosas/metabolismo , Regulación de la Expresión Génica , Silenciador del Gen , Hematopoyesis , Hiperlipidemias , Metabolismo de los Lípidos , Lípidos/química , Hígado/enzimología , Macrófagos/metabolismo , Ratones , Ratones Noqueados para ApoE , Células RAW 264.7
8.
Diabetes Care ; 39(1): 39-42, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26223240

RESUMEN

OBJECTIVE: Diet therapy in gestational diabetes mellitus (GDM) has focused on carbohydrate restriction but is poorly substantiated. In this pilot randomized clinical trial, we challenged the conventional low-carbohydrate/higher-fat (LC/CONV) diet, hypothesizing that a higher-complex carbohydrate/lower-fat (CHOICE) diet would improve maternal insulin resistance (IR), adipose tissue (AT) lipolysis, and infant adiposity. RESEARCH DESIGN AND METHODS: At 31 weeks, 12 diet-controlled overweight/obese women with GDM were randomized to an isocaloric LC/CONV (40% carbohydrate/45% fat/15% protein; n = 6) or CHOICE (60%/25%/15%; n = 6) diet. All meals were provided. AT was biopsied at 37 weeks. RESULTS: After ∼7 weeks, fasting glucose (P = 0.03) and free fatty acids (P = 0.06) decreased on CHOICE, whereas fasting glucose increased on LC/CONV (P = 0.03). Insulin suppression of AT lipolysis was improved on CHOICE versus LC/CONV (56 vs. 31%, P = 0.005), consistent with improved IR. AT expression of multiple proinflammatory genes was lower on CHOICE (P < 0.01). Infant adiposity trended lower with CHOICE (10.1 ± 1.4 vs. 12.6 ± 2%, respectively). CONCLUSIONS: A CHOICE diet may improve maternal IR and infant adiposity, challenging recommendations for a LC/CONV diet.


Asunto(s)
Diabetes Gestacional/dietoterapia , Dieta para Diabéticos , Dieta con Restricción de Grasas , Inflamación/epidemiología , Resistencia a la Insulina , Tejido Adiposo/metabolismo , Adulto , Glucemia/metabolismo , Ayuno , Ácidos Grasos no Esterificados/metabolismo , Femenino , Índice Glucémico , Humanos , Inflamación/sangre , Insulina/metabolismo , Obesidad/dietoterapia , Obesidad/metabolismo , Sobrepeso/dietoterapia , Proyectos Piloto , Embarazo , Adulto Joven
9.
PLoS One ; 9(1): e80949, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24465369

RESUMEN

Current estimates suggest that over one-third of the adult population has metabolic syndrome and three-fourths of the obese population has non-alcoholic fatty liver disease (NAFLD). Inflammation in metabolic tissues has emerged as a universal feature of obesity and its co-morbidities, including NAFLD. Natural Killer T (NKT) cells are a subset of innate immune cells that abundantly reside within the liver and are readily activated by lipid antigens. There is general consensus that NKT cells are pivotal regulators of inflammation; however, disagreement exists as to whether NKT cells exert pathogenic or suppressive functions in obesity. Here we demonstrate that CD1d(-/-) mice, which lack NKT cells, were more susceptible to weight gain and fatty liver following high fat diet (HFD) feeding. Compared with their WT counterparts, CD1d(-/-) mice displayed increased adiposity and greater induction of inflammatory genes in the liver suggestive of the precursors of NAFLD. Calorimetry studies revealed a significant increase in food intake and trends toward decreased metabolic rate and activity in CD1d(-/-) mice compared with WT mice. Based on these findings, our results suggest that NKT cells play a regulatory role that helps to prevent diet-induced obesity and metabolic dysfunction and may play an important role in mechanisms governing cross-talk between metabolism and the immune system to regulate energy balance and liver health.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Células T Asesinas Naturales/citología , Animales , Antígenos CD1d/genética , Antígenos CD1d/metabolismo , Calorimetría Indirecta , Femenino , Immunoblotting , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Células T Asesinas Naturales/fisiología , Obesidad/etiología , Obesidad/inmunología , Obesidad/metabolismo , ARN Mensajero/genética
10.
PLoS One ; 8(6): e67791, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23825686

RESUMEN

Maternal and pediatric obesity has risen dramatically over recent years, and is a known predictor of adverse long-term metabolic outcomes in offspring. However, which particular aspects of obese pregnancy promote such outcomes is less clear. While maternal obesity increases both maternal and placental inflammation, it is still unknown whether this is a dominant mechanism in fetal metabolic programming. In this study, we utilized the Fat-1 transgenic mouse to test whether increasing the maternal n-3/n-6 tissue fatty acid ratio could reduce the consequences of maternal obesity-associated inflammation and thereby mitigate downstream developmental programming. Eight-week-old WT or hemizygous Fat-1 C57BL/6J female mice were placed on a high-fat diet (HFD) or control diet (CD) for 8 weeks prior to mating with WT chow-fed males. Only WT offspring from Fat-1 mothers were analyzed. WT-HFD mothers demonstrated increased markers of infiltrating adipose tissue macrophages (P<0.02), and a striking increase in 12 serum pro-inflammatory cytokines (P<0.05), while Fat1-HFD mothers remained similar to WT-CD mothers, despite equal weight gain. E18.5 Fetuses from WT-HFD mothers had larger placentas (P<0.02), as well as increased placenta and fetal liver TG deposition (P<0.01 and P<0.02, respectively) and increased placental LPL TG-hydrolase activity (P<0.02), which correlated with degree of maternal insulin resistance (r = 0.59, P<0.02). The placentas and fetal livers from Fat1-HFD mothers were protected from this excess placental growth and fetal-placental lipid deposition. Importantly, maternal protection from excess inflammation corresponded with improved metabolic outcomes in adult WT offspring. While the offspring from WT-HFD mothers weaned onto CD demonstrated increased weight gain (P<0.05), body and liver fat (P<0.05 and P<0.001, respectively), and whole body insulin resistance (P<0.05), these were prevented in WT offspring from Fat1-HFD mothers. Our results suggest that reducing excess maternal inflammation may be a promising target for preventing adverse fetal metabolic outcomes in pregnancies complicated by maternal obesity.


Asunto(s)
Ácidos Grasos Omega-3/metabolismo , Ácidos Grasos Omega-6/metabolismo , Feto/metabolismo , Madres , Obesidad/metabolismo , Complicaciones del Embarazo/metabolismo , Animales , Dieta Alta en Grasa/efectos adversos , Femenino , Feto/embriología , Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Placenta/metabolismo , Embarazo
11.
J Biol Chem ; 287(41): 34349-60, 2012 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-22902781

RESUMEN

Strong evidence exists for a link between chronic low level inflammation and dietary-induced insulin resistance; however, little is known about the transcriptional networks involved. Here we show that high fat diet (HFD) or saturated fatty acid exposure directly activates CCAAT/enhancer-binding protein ß (C/EBPß) protein expression in liver, adipocytes, and macrophages. Global C/EBPß deletion prevented HFD-induced inflammation and surprisingly increased mitochondrial gene expression in white adipose tissue along with brown adipose tissue markers PRDM16, CIDEa, and UCP1, consistent with a resistance to HFD-induced obesity. In isolated peritoneal macrophages from C/EBPß(-/-) mice, the anti-inflammatory gene LXRα and its targets SCD1 and DGAT2 were strikingly up-regulated along with IL-10, while NLRP3, a gene important for activating the inflammasome, was suppressed in response to palmitate. Using RAW 264.7 macrophage cells or 3T3-L1 adipocytes, C/EBPß knockdown prevented palmitate-induced inflammation and p65-NFκB DNA binding activity, while C/EBPß overexpression induced NFκB binding, JNK activation, and pro-inflammatory cytokine gene expression directly. Finally, chimeric bone marrow mice transplanted with bone marrow lacking C/EBPß(-/-) demonstrated reduced systemic and adipose tissue inflammatory markers, macrophage content, and maintained insulin sensitivity on HFD. Taken together, these results demonstrate that HFD or palmitate exposure triggers C/EBPß expression that controls expression of distinct aspects of alternative macrophage activation. Reducing C/EBPß in macrophages confers protection from HFD-induced systemic inflammation and insulin resistance, suggesting it may be an attractive therapeutic target for ameliorating obesity-induced inflammatory responses.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/biosíntesis , Grasas de la Dieta/efectos adversos , Regulación de la Expresión Génica/efectos de los fármacos , Activación de Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Obesidad/metabolismo , Células 3T3-L1 , Tejido Adiposo Pardo/patología , Animales , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Grasas de la Dieta/farmacología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Inflamasomas/genética , Inflamasomas/metabolismo , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Interleucina-10/genética , Interleucina-10/metabolismo , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Activación de Macrófagos/genética , Macrófagos/patología , Ratones , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/patología , Proteína con Dominio Pirina 3 de la Familia NLR , Obesidad/inducido químicamente , Obesidad/genética , Obesidad/patología , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo
12.
Diabetes ; 58(1): 116-24, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18952837

RESUMEN

OBJECTIVE: Skeletal muscle-specific LPL knockout mouse (SMLPL(-/-)) were created to study the systemic impact of reduced lipoprotein lipid delivery in skeletal muscle on insulin sensitivity, body weight, and composition. RESEARCH DESIGN AND METHODS: Tissue-specific insulin sensitivity was assessed using a hyperinsulinemic-euglycemic clamp and 2-deoxyglucose uptake. Gene expression and insulin-signaling molecules were compared in skeletal muscle and liver of SMLPL(-/-) and control mice. RESULTS: Nine-week-old SMLPL(-/-) mice showed no differences in body weight, fat mass, or whole-body insulin sensitivity, but older SMLPL(-/-) mice had greater weight gain and whole-body insulin resistance. High-fat diet feeding accelerated the development of obesity. In young SMLPL(-/-) mice, insulin-stimulated glucose uptake was increased 58% in the skeletal muscle, but was reduced in white adipose tissue (WAT) and heart. Insulin action was also diminished in liver: 40% suppression of hepatic glucose production in SMLPL(-/-) vs. 90% in control mice. Skeletal muscle triglyceride was 38% lower, and insulin-stimulated phosphorylated Akt (Ser473) was twofold greater in SMLPL(-/-) mice without changes in IRS-1 tyrosine phosphorylation and phosphatidylinositol 3-kinase activity. Hepatic triglyceride and liver X receptor, carbohydrate response element-binding protein, and PEPCK mRNAs were unaffected in SMLPL(-/-) mice, but peroxisome proliferator-activated receptor (PPAR)-gamma coactivator-1alpha and interleukin-1beta mRNAs were higher, and stearoyl-coenzyme A desaturase-1 and PPARgamma mRNAs were reduced. CONCLUSIONS: LPL deletion in skeletal muscle reduces lipid storage and increases insulin signaling in skeletal muscle without changes in body composition. Moreover, lack of LPL in skeletal muscle results in insulin resistance in other key metabolic tissues and ultimately leads to obesity and systemic insulin resistance.


Asunto(s)
Resistencia a la Insulina/genética , Insulina/farmacología , Lipoproteína Lipasa/genética , Hígado/efectos de los fármacos , Absorciometría de Fotón , Animales , Glucemia/metabolismo , Composición Corporal , Citocinas/sangre , Técnica de Clampeo de la Glucosa , Prueba de Tolerancia a la Glucosa , Hipoglucemiantes/farmacología , Lípidos/sangre , Lipoproteína Lipasa/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Músculo Esquelético/enzimología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Transducción de Señal/fisiología
13.
J Cell Sci ; 118(Pt 16): 3653-62, 2005 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16046480

RESUMEN

Exocytotic incorporation of plasmalemmal precursor vesicles (PPVs) into the cell surface is necessary for axonal outgrowth and is known to occur mainly at the nerve growth cone. We have demonstrated recently that plasmalemmal expansion is regulated at the growth cone by IGF-1, but not by BDNF, in a manner that is quasi independent of the neuron's perikaryon. To begin elucidating the signaling pathway by which exocytosis of the plasmalemmal precursor is regulated, we studied activation of the IRS/PI3K/Akt pathway in isolated growth cones and hippocampal neurons in culture stimulated with IGF-1 or BDNF. Our results show that IGF-1, but not BDNF, significantly and rapidly stimulates IRS/PI3K/Akt and membrane expansion. Inhibition of PI3K with Wortmannin or LY294002 blocked IGF-1-stimulated plasmalemmal expansion at the growth cones of cultured neurons. Finally, our results show that, upon stimulation with IGF-1, most active PI3K becomes associated with distal microtubules in the proximal or central domain of the growth cone. Taken together, our results suggest a critical role for IGF-1 and the IRS/PI3K/Akt pathway in the process of membrane assembly at the axonal growth cone.


Asunto(s)
Membrana Celular/metabolismo , Sistema Nervioso Central/embriología , Exocitosis/fisiología , Conos de Crecimiento/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Membrana Celular/efectos de los fármacos , Células Cultivadas , Sistema Nervioso Central/citología , Sistema Nervioso Central/crecimiento & desarrollo , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Inhibidores Enzimáticos , Exocitosis/efectos de los fármacos , Conos de Crecimiento/efectos de los fármacos , Conos de Crecimiento/ultraestructura , Hipocampo/citología , Hipocampo/embriología , Hipocampo/crecimiento & desarrollo , Factor I del Crecimiento Similar a la Insulina/farmacología , Fusión de Membrana/efectos de los fármacos , Fusión de Membrana/fisiología , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , Ratas , Transducción de Señal/fisiología , Vesículas Transportadoras/efectos de los fármacos , Vesículas Transportadoras/metabolismo , Vesículas Transportadoras/ultraestructura
14.
J Neurosci ; 22(12): 4932-41, 2002 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-12077190

RESUMEN

Detection of a repellent factor, such as a semaphorin (Sema), causes localized collapse of the growth cone and directs the neurite away from the repellent. Growth cone collapse results from concomitant cytoskeletal rearrangements and detachment of adhesion sites from the extracellular matrix, via mostly unknown signaling mechanisms. In cultures of dorsal root ganglion neurons, we found that Sema3A treatment stimulates the synthesis of the eicosanoid, 12(S)-hydroxyeicosatetraenoic acid (HETE), whereas Sema3A-induced growth cone collapse is prevented when 12(S)-HETE synthesis is blocked with an inhibitor of 12/15-lipoxygenase (LO). Exogenously applied product of 12/15-LO, 12(S)-HETE, mimics Sema3A-induced collapse. As observed by interference reflection and confocal microscopy, 12(S)-HETE causes the loss of growth cone adhesion sites. The adhesion site effect seems partially independent of the actin cytoskeleton because growth cones treated with Sema3A and 12/15-LO inhibitor remain spread despite actin cytoskeleton loss. These studies demonstrate that 12/15-LO activity is a necessary step in Sema3A collapse signaling in growth cones and suggest a mechanism for its action.


Asunto(s)
Araquidonato 12-Lipooxigenasa/fisiología , Araquidonato 15-Lipooxigenasa/fisiología , Glicoproteínas/farmacología , Conos de Crecimiento/enzimología , Conos de Crecimiento/fisiología , Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico/biosíntesis , Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico/farmacología , Animales , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Conos de Crecimiento/efectos de los fármacos , Conos de Crecimiento/ultraestructura , Proteínas del Tejido Nervioso/análisis , Neuropilina-1 , Ratas , Ratas Sprague-Dawley , Semaforina-3A
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...