Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Commun Biol ; 6(1): 8, 2023 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-36599953

RESUMEN

Transmembrane protein 135 (TMEM135) is thought to participate in the cellular response to increased intracellular lipids yet no defined molecular function for TMEM135 in lipid metabolism has been identified. In this study, we performed a lipid analysis of tissues from Tmem135 mutant mice and found striking reductions of docosahexaenoic acid (DHA) across all Tmem135 mutant tissues, indicating a role of TMEM135 in the production of DHA. Since all enzymes required for DHA synthesis remain intact in Tmem135 mutant mice, we hypothesized that TMEM135 is involved in the export of DHA from peroxisomes. The Tmem135 mutation likely leads to the retention of DHA in peroxisomes, causing DHA to be degraded within peroxisomes by their beta-oxidation machinery. This may lead to generation or alteration of ligands required for the activation of peroxisome proliferator-activated receptor a (PPARa) signaling, which in turn could result in increased peroxisomal number and beta-oxidation enzymes observed in Tmem135 mutant mice. We confirmed this effect of PPARa signaling by detecting decreased peroxisomes and their proteins upon genetic ablation of Ppara in Tmem135 mutant mice. Using Tmem135 mutant mice, we also validated the protective effect of increased peroxisomes and peroxisomal beta-oxidation on the metabolic disease phenotypes of leptin mutant mice which has been observed in previous studies. Thus, we conclude that TMEM135 has a role in lipid homeostasis through its function in peroxisomes.


Asunto(s)
Ácidos Docosahexaenoicos , Metabolismo de los Lípidos , Proteínas de la Membrana , Peroxisomas , Animales , Ratones , Ácidos Docosahexaenoicos/metabolismo , Homeostasis , Oxidación-Reducción , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Peroxisomas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Mitocondriales/metabolismo
2.
Nat Metab ; 3(10): 1327-1341, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34663973

RESUMEN

Calorie restriction (CR) promotes healthy ageing in diverse species. Recently, it has been shown that fasting for a portion of each day has metabolic benefits and promotes lifespan. These findings complicate the interpretation of rodent CR studies, in which animals typically eat only once per day and rapidly consume their food, which collaterally imposes fasting. Here we show that a prolonged fast is necessary for key metabolic, molecular and geroprotective effects of a CR diet. Using a series of feeding regimens, we dissect the effects of calories and fasting, and proceed to demonstrate that fasting alone recapitulates many of the physiological and molecular effects of CR. Our results shed new light on how both when and how much we eat regulate metabolic health and longevity, and demonstrate that daily prolonged fasting, and not solely reduced caloric intake, is likely responsible for the metabolic and geroprotective benefits of a CR diet.


Asunto(s)
Envejecimiento/metabolismo , Restricción Calórica , Animales , Longevidad/fisiología , Ratones
3.
mSphere ; 5(3)2020 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-32581074

RESUMEN

The incidence of metabolic syndrome continues to rise globally. In mice, intravenous administration of interleukin-22 (IL-22) ameliorates various disease phenotypes associated with diet-induced metabolic syndrome. In patients, oral treatment is favored over intravenous treatment, but methodologies to deliver IL-22 via the oral route are nonexistent. The goal of this study was to assess to what extent engineered Lactobacillus reuteri secreting IL-22 could ameliorate nonalcoholic fatty liver disease. We used a mouse model of diet-induced obesity and assessed various markers of metabolic syndrome following treatment with L. reuteri and a recombinant derivative. Mice that received an 8-week treatment of wild-type probiotic gained less weight and had a smaller fat pad than the control group, but these phenotypes were not further enhanced by recombinant L. reuteri However, L. reuteri secreting IL-22 significantly reduced liver weight and triglycerides at levels that exceeded those of the probiotic wild-type treatment group. Our findings are interesting in light of the observed phenotypes associated with reduced nonalcoholic liver disease, in humans the most prevalent chronic liver disease, following treatment of a next-generation probiotic that is administered orally. Once biological and environmental containment strategies are in place, therapeutic applications of recombinant Lactobacillus reuteri are on the horizon.IMPORTANCE In humans, nonalcoholic fatty liver disease (NAFLD) is the most prevalent liver disease due to the increased prevalence of obesity. While treatment of NAFLD is often geared toward lifestyle changes, such as diet and exercise, the use of dietary supplements such as probiotics is underinvestigated. Here, we report that probiotic Lactobacillus reuteri reduces fatty liver in a mouse model of diet-induced obesity. This phenotype was further enhanced upon delivery of recombinant interleukin-22 by engineered Lactobacillus reuteri These observations pave the road to a better understanding of probiotic mechanisms driving the reduction of diet-induced steatosis and to development of next-generation probiotics for use in the clinic. Ultimately, these studies may lead to rational selection of (engineered) probiotics to ameliorate fatty liver disease.


Asunto(s)
Hígado Graso/prevención & control , Interleucinas/administración & dosificación , Limosilactobacillus reuteri/genética , Obesidad/terapia , Probióticos/uso terapéutico , Animales , Biomarcadores , Dieta , Modelos Animales de Enfermedad , Interleucinas/genética , Masculino , Síndrome Metabólico/terapia , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/genética , Interleucina-22
4.
PLoS One ; 13(6): e0199213, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29953483

RESUMEN

Although prenatal alcohol exposure (PAE) reduces offspring growth, it may increase obesity risk at adolescence. Animal models of PAE display glucose intolerance and increased adiposity, suggesting that PAE causes metabolic reprogramming. We tested this hypothesis in a mouse model of binge PAE, wherein pregnant C57Bl/6J females received 3 g/kg alcohol (ETOH) daily from gestational day 12.5 to 17.5; maltodextrin (MD) and medium chain triglycerides (MCT) served as isocaloric nutritional controls, and sham (H2O) treatment controlled for gavage stress. Our comprehensive assessment quantified body composition, energy expenditure, glucose tolerance, and cardiovascular function in offspring at age 17 weeks. Although ETOH pups were initially lighter than all other groups, they did not have a unique obesogenic phenotype. Instead, a similar obesogenic phenotype emerged in all three caloric groups (MCT, MD, ETOH), such that caloric groups had greater post-weaning weight gain (both sexes), reduced gonadal fat weight (males), and reduced glucose clearance (males) compared against H2O offspring. PAE did not affect body composition, respiratory exchange ratio, metabolic adaption to high-fat or low-fat diet, eating behavior, and blood pressure, and ETOH values did not differ from those obtained from isocaloric controls. Exposure to a higher alcohol dose (4.5 g/kg) or a high-fat (60%) diet did not exacerbate differences in body composition or glucose tolerance. "PAE-specific" effects on postnatal growth, glucose tolerance, adiposity, or hypertension only emerged when PAE offspring were compared just against H2O controls, or against MD controls. We conclude that prior reports of obesity and glucose intolerance in adult PAE offspring reflect the contribution of added gestational calories, and not alcohol's pharmacologic action. Results suggest that the increased adiposity risk in FASD is not caused by metabolic reprogramming, and instead originates from behavioral, medication, and/or dietary practices. This study highlights the importance of appropriate dietary controls in nutritional studies of PAE.


Asunto(s)
Consumo de Bebidas Alcohólicas/efectos adversos , Trastornos del Espectro Alcohólico Fetal/etiología , Exposición Materna , Síndrome Metabólico/etiología , Síndrome Metabólico/metabolismo , Efectos Tardíos de la Exposición Prenatal , Adiposidad , Animales , Biomarcadores , Peso Corporal , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Metabolismo Energético , Conducta Alimentaria , Femenino , Desarrollo Fetal/efectos de los fármacos , Intolerancia a la Glucosa , Hemodinámica , Masculino , Ratones , Embarazo
5.
J Lipid Res ; 58(6): 1091-1099, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28420705

RESUMEN

In mammals, ether lipids exert a wide spectrum of signaling and structural functions, such as stimulation of immune responses, anti-tumor activities, and enhancement of sperm functions. Abnormal accumulation of monoalkyl-diacylglycerol (MADAG) was found in Wolman's disease, a human genetic disorder defined by a deficiency in lysosomal acid lipase. In the current study, we found that among the nine recombinant human lipid acyltransferases examined, acyl-CoA:diacylglycerol acyltransferase (DGAT)1, DGAT2, acyl-CoA:monoacylglycerol acyltransferase (MGAT)2, MGAT3, acyl-CoA:wax-alcohol acyltransferase 2/MFAT, and DGAT candidate 3 were able to use 1-monoalkylglycerol (1-MAkG) as an acyl acceptor for the synthesis of monoalkyl-monoacylglycerol (MAMAG). These enzymes demonstrated different enzymatic turnover rates and relative efficiencies for the first and second acylation steps leading to the synthesis of MAMAG and MADAG, respectively. They also exhibited different degrees of substrate preference when presented with 1-monooleoylglycerol versus 1-MAkG. In CHO-K1 cells, treatment with DGAT1 selective inhibitor, XP-620, completely blocked the synthesis of MADAG, indicating that DGAT1 is the predominant enzyme responsible for the intracellular synthesis of MADAG in this model system. The levels of MADAG in the adrenal gland of DGAT1 KO mice were reduced as compared with those of the WT mice, suggesting that DGAT1 is a major enzyme for the synthesis of MADAG in this tissue. Our findings indicate that several of these lipid acyltransferases may be able to synthesize neutral ether lipids in mammals.


Asunto(s)
Aciltransferasas/metabolismo , Diglicéridos/biosíntesis , Diglicéridos/química , Éteres/química , Acilación , Animales , Células CHO , Chlorocebus aethiops , Cricetulus , Diglicéridos/metabolismo , Humanos
6.
Nutr Metab (Lond) ; 13: 48, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27478484

RESUMEN

BACKGROUND: Intestinal absorption of dietary lipids involves their hydrolysis in the lumen of proximal intestine as well as uptake, intracellular transport and re-assembly of hydrolyzed lipids in enterocytes, leading to the formation and secretion of the lipoproteins chylomicrons and HDL. In this study, we examined the potential involvement of cytosolic lipid droplets (CLD) whose function in the process of lipid absorption is poorly understood. METHODS: Intestinal lipid absorption was studied in mouse after gavage. Three populations of CLD were purified by density ultracentrifugations, as well as the brush border membranes, which were analyzed by western-blots. Immunofluorescent localization of membranes transporters or metabolic enzymes, as well as kinetics of CLD production, were also studied in intestine or Caco-2 cells. RESULTS: We isolated three populations of CLD (ranging from 15 to 1000 nm) which showed differential expression of the major lipid transporters scavenger receptor BI (SR-BI), cluster of differentiation 36 (CD-36), Niemann Pick C-like 1 (NPC1L1), and the ATP-binding cassette transporters ABCG5/G8 but also caveolin 2 and fatty acid binding proteins. The enzyme monoacylglycerol acyltransferase 2 (MGAT2) was identified in the brush border membrane (BBM) in addition to the endoplasmic reticulum, suggesting local synthesis of triglycerides and CLD at both places. CONCLUSIONS: We show a very fast production of CLD by enterocytes associated with a transfer of apical constituents as lipid transporters. Our findings suggest that following their uptake by enterocytes, lipids can be partially metabolized at the BBM and packaged into CLD for their transportation to the ER.

7.
J Lipid Res ; 57(4): 616-30, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26880786

RESUMEN

Reducing triacylglycerol (TAG) in the liver continues to pose a challenge in states of nonalcoholic hepatic steatosis. MonoacylglycerolO-acyltransferase (MOGAT) enzymes convert monoacylglycerol (MAG) to diacylglycerol, a precursor for TAG synthesis, and are involved in a major pathway of TAG synthesis in selected tissues, such as small intestine. MOGAT1 possesses MGAT activity in in vitro assays, but its physiological function in TAG metabolism is unknown. Recent studies suggest a role for MOGAT1 in hepatic steatosis in lipodystrophic [1-acylglycerol-3-phosphateO-acyltransferase (Agpat)2(-/-)] and obese (ob/ob) mice. To test this, we deletedMogat1in theAgpat2(-/-)andob/obgenetic background to generateMogat1(-/-);Agpat2(-/-)andMogat1(-/-);ob/obdouble knockout (DKO) mice. Here we report that, despite the absence ofMogat1in either DKO mouse model, we did not find any decrease in liver TAG by 16 weeks of age. Additionally, there were no measureable changes in plasma glucose (diabetes) and insulin resistance. Our data indicate a minimal role, if any, of MOGAT1 in liver TAG synthesis, and that TAG synthesis in steatosis associated with lipodystrophy and obesity is independent of MOGAT1. Our findings suggest that MOGAT1 likely has an alternative function in vivo.


Asunto(s)
Aciltransferasas/deficiencia , Aciltransferasas/genética , Eliminación de Gen , Lipodistrofia/genética , Enfermedad del Hígado Graso no Alcohólico/enzimología , Enfermedad del Hígado Graso no Alcohólico/genética , Animales , Femenino , Insulina/sangre , Resistencia a la Insulina , Lipodistrofia/complicaciones , Lipodistrofia/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Obesos , Enfermedad del Hígado Graso no Alcohólico/sangre , Enfermedad del Hígado Graso no Alcohólico/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
8.
J Lipid Res ; 56(3): 489-501, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25231105

RESUMEN

The intestine plays a prominent role in the biosynthesis of triacylglycerol (triglyceride; TAG). Digested dietary TAG is repackaged in the intestine to form the hydrophobic core of chylomicrons, which deliver metabolic fuels, essential fatty acids, and other lipid-soluble nutrients to the peripheral tissues. By controlling the flux of dietary fat into the circulation, intestinal TAG synthesis can greatly impact systemic metabolism. Genes encoding many of the enzymes involved in TAG synthesis have been identified. Among TAG synthesis enzymes, acyl-CoA:monoacylglycerol acyltransferase 2 and acyl-CoA:diacylglycerol acyltransferase (DGAT)1 are highly expressed in the intestine. Their physiological functions have been examined in the context of whole organisms using genetically engineered mice and, in the case of DGAT1, specific inhibitors. An emerging theme from recent findings is that limiting the rate of TAG synthesis in the intestine can modulate gut hormone secretion, lipid metabolism, and systemic energy balance. The underlying mechanisms and their implications for humans are yet to be explored. Pharmacological inhibition of TAG hydrolysis in the intestinal lumen has been employed to combat obesity and associated disorders with modest efficacy and unwanted side effects. The therapeutic potential of inhibiting specific enzymes involved in intestinal TAG synthesis warrants further investigation.


Asunto(s)
Metabolismo Energético/fisiología , Absorción Intestinal/fisiología , Mucosa Intestinal/metabolismo , Metabolismo de los Lípidos/fisiología , Triglicéridos/biosíntesis , Animales , Quilomicrones/genética , Quilomicrones/metabolismo , Diacilglicerol O-Acetiltransferasa/biosíntesis , Diacilglicerol O-Acetiltransferasa/genética , Regulación Enzimológica de la Expresión Génica/fisiología , Humanos , Ratones , Triglicéridos/genética
9.
PLoS Genet ; 10(8): e1004514, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25101993

RESUMEN

Homeostatic temperature regulation is fundamental to mammalian physiology and is controlled by acute and chronic responses of local, endocrine and nervous regulators. Here, we report that loss of the heparan sulfate proteoglycan, syndecan-1, causes a profoundly depleted intradermal fat layer, which provides crucial thermogenic insulation for mammals. Mice without syndecan-1 enter torpor upon fasting and show multiple indicators of cold stress, including activation of the stress checkpoint p38α in brown adipose tissue, liver and lung. The metabolic phenotype in mutant mice, including reduced liver glycogen, is rescued by housing at thermoneutrality, suggesting that reduced insulation in cool temperatures underlies the observed phenotypes. We find that syndecan-1, which functions as a facultative lipoprotein uptake receptor, is required for adipocyte differentiation in vitro. Intradermal fat shows highly dynamic differentiation, continuously expanding and involuting in response to hair cycle and ambient temperature. This physiology probably confers a unique role for Sdc1 in this adipocyte sub-type. The PPARγ agonist rosiglitazone rescues Sdc1-/- intradermal adipose tissue, placing PPARγ downstream of Sdc1 in triggering adipocyte differentiation. Our study indicates that disruption of intradermal adipose tissue development results in cold stress and complex metabolic pathology.


Asunto(s)
Diferenciación Celular/genética , Proteína Quinasa 14 Activada por Mitógenos/genética , PPAR gamma/genética , Estrés Fisiológico/genética , Sindecano-1/genética , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Animales , Frío , Ratones , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , PPAR gamma/agonistas , PPAR gamma/metabolismo , Rosiglitazona , Sindecano-1/metabolismo , Tiazolidinedionas/administración & dosificación
10.
J Biol Chem ; 289(25): 17338-49, 2014 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-24784138

RESUMEN

The absorption of dietary fat involves the re-esterification of digested triacylglycerol in the enterocytes, a process catalyzed by acyl-CoA:monoacylglycerol acyltransferase (MGAT) 2. Mice without a functional gene encoding MGAT2 (Mogat2(-/-)) are protected from diet-induced obesity. Surprisingly, these mice absorb normal amounts of dietary fat but increase their energy expenditure. MGAT2 is expressed in tissues besides intestine, including adipose tissue in both mice and humans. To test the hypothesis that intestinal MGAT2 regulates systemic energy balance, we generated and characterized mice deficient in MGAT2 specifically in the small intestine (Mogat2(IKO)). We found that, like Mogat2(-/-) mice, Mogat2(IKO) mice also showed a delay in fat absorption, a decrease in food intake, and a propensity to use fatty acids as fuel when first exposed to a high fat diet. Mogat2(IKO) mice increased energy expenditure although to a lesser degree than Mogat2(-/-) mice and were protected against diet-induced weight gain and associated comorbidities, including hepatic steatosis, hypercholesterolemia, and glucose intolerance. These findings illustrate that intestinal lipid metabolism plays a crucial role in the regulation of systemic energy balance and may be a feasible intervention target. In addition, they suggest that MGAT activity in extraintestinal tissues may also modulate energy metabolism.


Asunto(s)
Grasas de la Dieta/efectos adversos , Intolerancia a la Glucosa/enzimología , Intestinos/enzimología , N-Acetilglucosaminiltransferasas/metabolismo , Obesidad/enzimología , Animales , Grasas de la Dieta/farmacología , Ingestión de Alimentos/genética , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Hígado Graso/enzimología , Hígado Graso/genética , Hígado Graso/patología , Hígado Graso/prevención & control , Eliminación de Gen , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/patología , Intolerancia a la Glucosa/prevención & control , Humanos , Hipercolesterolemia/enzimología , Hipercolesterolemia/genética , Hipercolesterolemia/patología , Hipercolesterolemia/prevención & control , Absorción Intestinal/efectos de los fármacos , Absorción Intestinal/genética , Intestinos/patología , Ratones , Ratones Noqueados , N-Acetilglucosaminiltransferasas/genética , Obesidad/inducido químicamente , Obesidad/genética , Obesidad/patología , Obesidad/prevención & control
11.
J Lipid Res ; 54(6): 1644-1652, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23536640

RESUMEN

Acyl CoA:monoacylglycerol acyltransferase (MGAT) catalyzes the resynthesis of triacylglycerol, a crucial step in the absorption of dietary fat. Mice lacking the gene Mogat2, which codes for an MGAT highly expressed in the small intestine, are resistant to obesity and other metabolic disorders induced by high-fat feeding. Interestingly, these Mogat2⁻/⁻ mice absorb normal amounts of dietary fat but exhibit a reduced rate of fat absorption, increased energy expenditure, decreased respiratory exchange ratio, and impaired metabolic efficiency. MGAT2 is expressed in tissues besides intestine. To test the hypothesis that intestinal MGAT2 enhances metabolic efficiency and promotes the storage of metabolic fuels, we introduced the human MOGAT2 gene driven by the intestine-specific villin promoter into Mogat2⁻/⁻ mice. We found that the expression of MOGAT2 in the intestine increased intestinal MGAT activity, restored fat absorption rate, partially corrected energy expenditure, and promoted weight gain upon high-fat feeding. However, the changes in respiratory exchange ratio were not reverted, and the recoveries in metabolic efficiency and weight gain were incomplete. These data indicate that MGAT2 in the intestine plays an indispensable role in enhancing metabolic efficiency but also raise the possibility that MGAT2 in other tissues may contribute to the regulation of energy metabolism.


Asunto(s)
Aciltransferasas/biosíntesis , Metabolismo Energético , Regulación Enzimológica de la Expresión Génica , Absorción Intestinal/fisiología , Intestinos/enzimología , Aciltransferasas/genética , Animales , Grasas de la Dieta/metabolismo , Grasas de la Dieta/farmacología , Humanos , Ratones , Ratones Noqueados , Especificidad de Órganos/fisiología
12.
Am J Physiol Endocrinol Metab ; 302(7): E885-95, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22297302

RESUMEN

Phenylketonuria (PKU) is caused by a mutation in the phenylalanine (phe) hydroxylase gene and requires a low-phe diet plus amino acid (AA) formula to prevent cognitive impairment. Glycomacropeptide (GMP) contains minimal phe and provides a palatable alternative to AA formula. Our objective was to compare growth, body composition, and energy balance in Pah(enu2) (PKU) and wild-type mice fed low-phe GMP, low-phe AA, or high-phe casein diets from 3-23 wk of age. The 2 × 2 × 3 design included main effects of genotype, sex, and diet. Fat and lean mass were assessed by dual-energy X-ray absorptiometry, and acute energy balance was assessed by indirect calorimetry. PKU mice showed growth and lean mass similar to wild-type littermates fed the GMP or AA diets; however, they exhibited a 3-15% increase in energy expenditure, as reflected in oxygen consumption, and a 3-30% increase in food intake. The GMP diet significantly reduced energy expenditure, food intake, and plasma phe concentration in PKU mice compared with the casein diet. The high-phe casein diet or the low-phe AA diet induced metabolic stress in PKU mice, as reflected in increased energy expenditure and intake of food and water, increased renal and spleen mass, and elevated plasma cytokine concentrations consistent with systemic inflammation. The low-phe GMP diet significantly attenuated these adverse effects. Moreover, total fat mass, %body fat, and the respiratory exchange ratio (CO(2) produced/O(2) consumed) were significantly lower in PKU mice fed GMP compared with AA diets. In summary, GMP provides a physiological source of low-phe dietary protein that promotes growth and attenuates the metabolic stress induced by a high-phe casein or low-phe AA diet in PKU mice.


Asunto(s)
Caseínas/farmacología , Queso/análisis , Fragmentos de Péptidos/farmacología , Fenilcetonurias/tratamiento farmacológico , Fenilcetonurias/metabolismo , Absorciometría de Fotón , Aminoácidos/sangre , Aminoácidos/farmacología , Animales , Composición Corporal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Citocinas/sangre , Dieta , Dieta con Restricción de Proteínas , Metabolismo Energético/efectos de los fármacos , Crecimiento/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Tamaño de los Órganos/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Fenotipo , Fenilcetonurias/genética , Bazo/efectos de los fármacos , Bazo/crecimiento & desarrollo , Bazo/patología , Esplenomegalia/inducido químicamente , Esplenomegalia/patología
13.
PLoS One ; 6(12): e29111, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22195001

RESUMEN

Hibernating mammals cease feeding during the winter and rely primarily on stored lipids to fuel alternating periods of torpor and arousal. How hibernators manage large fluxes of lipids and sterols over the annual hibernation cycle is poorly understood. The aim of this study was to investigate lipid and cholesterol transport and storage in ground squirrels studied in spring, summer, and several hibernation states. Cholesterol levels in total plasma, HDL and LDL particles were elevated in hibernators compared with spring or summer squirrels. Hibernation increased plasma apolipoprotein A-I expression and HDL particle size. Expression of cholesterol 7 alpha-hydroxylase was 13-fold lower in hibernators than in active season squirrels. Plasma triglycerides were reduced by fasting in spring but not summer squirrels. In hibernators plasma ß-hydroxybutyrate was elevated during torpor whereas triglycerides were low relative to normothermic states. We conclude that the switch to a lipid-based metabolism during winter, coupled with reduced capacity to excrete cholesterol creates a closed system in which efficient use of lipoproteins is essential for survival.


Asunto(s)
Colesterol/sangre , Hibernación/fisiología , Lipoproteínas/sangre , Sciuridae/sangre , Sciuridae/fisiología , Ácido 3-Hidroxibutírico/sangre , Aciltransferasas/metabolismo , Animales , Hidrocarburo de Aril Hidroxilasas/genética , Hidrocarburo de Aril Hidroxilasas/metabolismo , Bilis/metabolismo , Temperatura Corporal/fisiología , Ácidos Grasos/sangre , Femenino , Regulación de la Expresión Génica , Hibernación/genética , Hidroximetilglutaril-CoA Reductasas/genética , Hidroximetilglutaril-CoA Reductasas/metabolismo , Masculino , Modelos Biológicos , Especificidad de Órganos , Sciuridae/genética , Estaciones del Año , Esteroide Hidroxilasas/genética , Esteroide Hidroxilasas/metabolismo , Triglicéridos/sangre
14.
J Lipid Res ; 52(9): 1723-32, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21734185

RESUMEN

Acyl CoA:monoacylglycerol acyltransferase 2 (MGAT2) is thought to be crucial for dietary fat absorption. Indeed, mice lacking the enzyme (Mogat2(-/-)) are resistant to obesity and other metabolic disorders induced by high-fat feeding. However, these mice absorb normal quantities of fat. To explore whether a high level of dietary fat is an essential part of the underlying mechanism(s), we examined metabolic responses of Mogat2(-/-) mice to diets containing varying levels of fat. Mogat2(-/-) mice exhibited 10-15% increases in energy expenditure compared with wild-type littermates; although high levels of dietary fat exacerbated the effect, this phenotype was expressed even on a fat-free diet. When deprived of food, Mogat2(-/-) mice expended energy and lost weight like wild-type controls. To determine whether MGAT2 deficiency protects against obesity in the absence of high-fat feeding, we crossed Mogat2(-/-) mice with genetically obese Agouti mice. MGAT2 deficiency increased energy expenditure and prevented these mice from gaining excess weight. Our results suggest that MGAT2 modulates energy expenditure through multiple mechanisms, including one independent of dietary fat; these findings also raise the prospect of inhibiting MGAT2 as a strategy for combating obesity and related metabolic disorders resulting from excessive calorie intake.


Asunto(s)
Aciltransferasas/deficiencia , Grasas de la Dieta/metabolismo , Metabolismo Energético , Ratones Obesos/metabolismo , Aumento de Peso , Aciltransferasas/genética , Animales , Ingestión de Alimentos/fisiología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Consumo de Oxígeno
15.
Nat Med ; 15(4): 442-6, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19287392

RESUMEN

Animals are remarkably efficient in absorbing dietary fat and assimilating this energy-dense nutrient into the white adipose tissue (WAT) for storage. Although this metabolic efficiency may confer an advantage in times of calorie deprivation, it contributes to obesity and associated metabolic disorders when dietary fat is abundant. Here we show that the intestinal lipid synthesis enzyme acyl CoA:monoacylglycerol acyltransferase-2 (MGAT2) has a crucial role in the assimilation of dietary fat and the accretion of body fat in mice. Mice lacking MGAT2 have a normal phenotype on a low-fat diet. However, on a high-fat diet, MGAT2-deficient mice are protected against developing obesity, glucose intolerance, hypercholesterolemia and fatty livers. Caloric intake is normal in MGAT2-deficient mice, and dietary fat is absorbed fully. However, entry of dietary fat into the circulation occurs at a reduced rate. This altered kinetics of fat absorption apparently results in more partitioning of dietary fat toward energy dissipation rather than toward storage in the WAT. Thus, our studies identify MGAT2 as a key determinant of energy metabolism in response to dietary fat and suggest that the inhibition of this enzyme may prove to be a useful strategy for treating obesity and other metabolic diseases associated with excessive fat intake.


Asunto(s)
Aciltransferasas/deficiencia , Grasas de la Dieta/efectos adversos , Obesidad/genética , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/fisiopatología , Animales , Temperatura Corporal , Peso Corporal , Ingestión de Energía , Metabolismo Energético , Femenino , Crecimiento , Hipercolesterolemia/prevención & control , Masculino , Ratones , Obesidad/prevención & control
16.
J Lipid Res ; 49(11): 2283-301, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18757836

RESUMEN

Triacylglycerols (triglycerides) (TGs) are the major storage molecules of metabolic energy and FAs in most living organisms. Excessive accumulation of TGs, however, is associated with human diseases, such as obesity, diabetes mellitus, and steatohepatitis. The final and the only committed step in the biosynthesis of TGs is catalyzed by acyl-CoA:diacylglycerol acyltransferase (DGAT) enzymes. The genes encoding two DGAT enzymes, DGAT1 and DGAT2, were identified in the past decade, and the use of molecular tools, including mice deficient in either enzyme, has shed light on their functions. Although DGAT enzymes are involved in TG synthesis, they have distinct protein sequences and differ in their biochemical, cellular, and physiological functions. Both enzymes may be useful as therapeutic targets for diseases. Here we review the current knowledge of DGAT enzymes, focusing on new advances since the cloning of their genes, including possible roles in human health and diseases.


Asunto(s)
Diacilglicerol O-Acetiltransferasa/fisiología , Glicerol/metabolismo , Lipogénesis/fisiología , Triglicéridos/biosíntesis , Animales , Diacilglicerol O-Acetiltransferasa/química , Diacilglicerol O-Acetiltransferasa/deficiencia , Diacilglicerol O-Acetiltransferasa/genética , Glicerol/química , Humanos , Isoenzimas/química , Isoenzimas/deficiencia , Isoenzimas/genética , Isoenzimas/fisiología , Lipogénesis/genética , Triglicéridos/química , Triglicéridos/genética
17.
Cell Metab ; 3(5): 305-7, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16679288

RESUMEN

The hydrolysis of fat stored in adipose tissues is crucial for providing energy during fasting and exercise, and dysregulation of fat breakdown may contribute to metabolic disease. In this issue of Cell Metabolism, report that CGI-58/ABHD5, a lipid-droplet-associated protein that is mutated in a rare disease characterized by excess lipid storage, activates adipose triglyceride lipase and thus may regulate fat mobilization.


Asunto(s)
Tejido Adiposo/enzimología , Esterasas/metabolismo , Lipasa/metabolismo , Errores Innatos del Metabolismo Lipídico/enzimología , Fosfolipasas A/metabolismo , Triglicéridos/metabolismo , 1-Acilglicerol-3-Fosfato O-Aciltransferasa , Animales , Activación Enzimática , Esterasas/genética , Humanos , Lipasa/genética , Errores Innatos del Metabolismo Lipídico/genética , Lipólisis , Mutación , Síndrome
18.
J Lipid Res ; 46(11): 2388-97, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16106050

RESUMEN

Acyl-CoA-dependent O-acyltransferases catalyze reactions in which fatty acyl-CoAs are joined to acyl acceptors containing free hydroxyl groups to produce neutral lipids. In this report, we characterize a human multifunctional O-acyltransferase (designated MFAT) that belongs to the acyl-CoA:diacylglycerol acyltransferase 2/acyl-CoA:monoacylglycerol acyltransferase (MGAT) gene family and is highly expressed in the skin. Membranes of insect cells and homogenates of mammalian cells overexpressing MFAT exhibited significantly increased MGAT, acyl-CoA:fatty acyl alcohol acyltransferase (wax synthase), and acyl-CoA:retinol acyltransferase (ARAT) activities, which catalyze the synthesis of diacylglycerols, wax monoesters, and retinyl esters, respectively. Furthermore, when provided with the appropriate substrates, intact mammalian cells overexpressing MFAT accumulated more waxes and retinyl esters than control cells. We conclude that MFAT is a multifunctional acyltransferase that likely plays an important role in lipid metabolism in human skin.


Asunto(s)
Aciltransferasas/química , Aciltransferasas/fisiología , Ésteres/química , Glicéridos/química , Piel/metabolismo , Animales , Northern Blotting , Células COS , Catálisis , Línea Celular , Chlorocebus aethiops , Clonación Molecular , ADN Complementario/metabolismo , Diacilglicerol O-Acetiltransferasa/química , Relación Dosis-Respuesta a Droga , Humanos , Immunoblotting , Insectos , Metabolismo de los Lípidos , Lípidos/química , Filogenia , Retinol O-Graso-Aciltransferasa/química , Distribución Tisular , Ceras
19.
J Lipid Res ; 46(7): 1502-11, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15834126

RESUMEN

The final step of triacylglycerol biosynthesis is catalyzed by acyl CoA:diacylglycerol acyltransferase (DGAT) enzymes. The two known DGATs, DGAT1 and DGAT2, are encoded by unrelated genes. Although both DGAT1 and DGAT2 knockout mice have reduced tissue triacylglycerol contents, they have disparate phenotypes, prompting us to investigate whether the two enzymes have unrecognized functional differences. We now report that DGAT1 exhibits additional acyltransferase activities in vitro, including those of acyl CoA:monoacylglycerol acyltransferase (MGAT), wax monoester and wax diester synthases, and acyl CoA:retinol acyltransferase (ARAT), which catalyze the synthesis of diacylglycerols, wax esters, and retinyl esters, respectively. These activities were demonstrated in in vitro assays with membranes from insect cells or homogenates from COS7 cells overexpressing DGAT1. Wax synthase and ARAT activities were also demonstrated in intact COS7 cells expressing DGAT1. Additionally, cells and tissues from DGAT1-deficient mice exhibited reduced ARAT activity, and the mice had increased levels of unesterified retinol in their livers on a high-retinol diet. Our findings indicate that DGAT1 can utilize a variety of acyl acceptors as substrates in vitro and suggest that these activities may be relevant to the in vivo functions of DGAT1.


Asunto(s)
Aciltransferasas/metabolismo , Diglicéridos/biosíntesis , Triglicéridos/biosíntesis , Vitamina A/metabolismo , Ceras/metabolismo , Animales , Células COS , Chlorocebus aethiops , Diacilglicerol O-Acetiltransferasa , Ratones , Spodoptera
20.
J Biol Chem ; 278(20): 18532-7, 2003 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-12621063

RESUMEN

Acyl CoA:monoacylglycerol acyltransferase (MGAT) catalyzes the synthesis of diacylglycerol, a precursor of triacylglycerol. In the intestine, MGAT plays a major role in the absorption of dietary fat by catalyzing the resynthesis of triacylglycerol in enterocytes. This resynthesis is required for the assembly of lipoproteins that transport absorbed fat to other tissues. Despite intense efforts, a gene encoding an intestinal MGAT has not been found. Previously, we identified a gene encoding MGAT1, which in mice is expressed in the stomach, kidney, adipose tissue, and liver but not in the intestine. We now report the identification of homologous genes in humans and mice encoding MGAT2. Expression of the MGAT2 cDNA in either insect or mammalian cells markedly increased MGAT activity in cell membranes. MGAT activity was proportional to the level of MGAT2 protein expressed, and the amount of diacylglycerol produced depended on the concentration of MGAT substrates (fatty acyl CoA or monoacylglycerol). In humans, the MGAT2 gene is highly expressed in the small intestine, liver, stomach, kidney, colon, and white adipose tissue; in mice, it is expressed predominantly in the small intestine. The discovery of the MGAT2 gene will facilitate studies to determine the functional role of MGAT2 in fat absorption in the intestine and to determine whether blocking MGAT activity in enterocytes is a feasible approach to inhibit fat absorption and treat obesity.


Asunto(s)
N-Acetilglucosaminiltransferasas/biosíntesis , N-Acetilglucosaminiltransferasas/química , Secuencia de Aminoácidos , Animales , Células COS , Línea Celular , Clonación Molecular , ADN Complementario/metabolismo , Diglicéridos/metabolismo , Relación Dosis-Respuesta a Droga , Enterocitos/metabolismo , Epítopos , Humanos , Insectos , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Ratones , Modelos Biológicos , Datos de Secuencia Molecular , N-Acetilglucosaminiltransferasas/fisiología , Homología de Secuencia de Aminoácido , Especificidad por Sustrato , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...