Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Endocrinol Metab ; 319(1): E133-E145, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32459527

RESUMEN

Bromocriptine mesylate treatment was examined in dogs fed a high fat diet (HFD) for 8 wk. After 4 wk on HFD, daily bromocriptine (Bromo; n = 6) or vehicle (CTR; n = 5) injections were administered. Oral glucose tolerance tests were performed before beginning HFD (OGTT1), 4 wk after HFD began (Bromo only), and after 7.5 wk on HFD (OGTT3). After 8 wk on HFD, clamp studies were performed, with infusion of somatostatin and intraportal replacement of insulin (4× basal) and glucagon (basal). From 0 to 90 min (P1), glucose was infused via peripheral vein to double the hepatic glucose load; and from 90 to 180 min (P2), glucose was infused via the hepatic portal vein at 4 mg·kg-1·min-1, with the HGL maintained at 2× basal. Bromo decreased the OGTT glucose ΔAUC0-30 and ΔAUC0-120 by 62 and 27%, respectively, P < 0.05 for both) without significantly altering the insulin response. Bromo dogs exhibited enhanced net hepatic glucose uptake (NHGU) compared with CTR (~33 and 21% greater, P1 and P2, respectively, P < 0.05). Nonhepatic glucose uptake (non-HGU) was increased ~38% in Bromo in P2 (P < 0.05). Bromo vs. CTR had higher (P < 0.05) rates of glucose infusion (36 and 30%) and non-HGU (~40 and 27%) than CTR during P1 and P2, respectively. In Bromo vs. CTR, hepatic 18:0/16:0 and 16:1/16:0 ratios tended to be elevated in triglycerides and were higher (P < 0.05) in phospholipids, consistent with a beneficial effect of bromocriptine on liver fat accumulation. Thus, bromocriptine treatment improved glucose disposal in a glucose-intolerant model, enhancing both NHGU and non-HGU.


Asunto(s)
Glucemia/efectos de los fármacos , Bromocriptina/farmacología , Dieta Alta en Grasa , Agonistas de Dopamina/farmacología , Intolerancia a la Glucosa/metabolismo , Hígado/efectos de los fármacos , Animales , Glucemia/metabolismo , Perros , Ácidos Grasos no Esterificados/metabolismo , Glucagón/efectos de los fármacos , Glucagón/metabolismo , Glucosa/metabolismo , Técnica de Clampeo de la Glucosa , Prueba de Tolerancia a la Glucosa , Glucógeno/metabolismo , Venas Hepáticas , Insulina/metabolismo , Ácido Láctico/metabolismo , Hígado/metabolismo , Vena Porta , Somatostatina
2.
Br J Nutr ; 117(11): 1615-1622, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28660850

RESUMEN

Dietary intake of PUFA has been associated with colorectal neoplasm risk; however, results from observational studies have been inconsistent. Most prior studies have utilised self-reported dietary measures to assess fatty acid exposure which might be more susceptible to measurement error and biases compared with biomarkers. The purpose of this study was to determine whether erythrocyte phospholipid membrane PUFA percentages are associated with colorectal adenoma risk. We included data from 904 adenoma cases and 835 polyp-free controls who participated in the Tennessee Colorectal Polyp Study, a large colonoscopy-based case-control study. Erythrocyte membrane PUFA percentages were measured using GC. Conditional logistic regression was used to calculate adjusted OR for risk of colorectal adenomas with erythrocyte membrane PUFA. Higher erythrocyte membrane percentages of arachidonic acid was associated with an increased risk of colorectal adenomas (adjusted OR 1·66; 95 % CI 1·05, 2·62, P trend=0·02) comparing the highest tertile to the lowest tertile. The effect size for arachidonic acid was more pronounced when restricting the analysis to advanced adenomas only. Higher erythrocyte membrane EPA percentages were associated with a trend towards a reduced risk of advanced colorectal adenomas (P trend=0·05). Erythrocyte membrane arachidonic acid percentages are associated with an increased risk of colorectal adenomas.


Asunto(s)
Adenoma/sangre , Ácido Araquidónico/sangre , Neoplasias Colorrectales/sangre , Ácido Eicosapentaenoico/sangre , Membrana Eritrocítica/metabolismo , Fosfolípidos/química , Adenoma/etiología , Adenoma/prevención & control , Biomarcadores/sangre , Estudios de Casos y Controles , Neoplasias Colorrectales/etiología , Neoplasias Colorrectales/prevención & control , Dieta , Femenino , Humanos , Modelos Logísticos , Masculino , Persona de Mediana Edad , Oportunidad Relativa , Fosfolípidos/sangre , Factores de Riesgo , Tennessee
3.
Am J Physiol Endocrinol Metab ; 299(4): E607-14, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20663988

RESUMEN

Hepatic glucagon action increases in response to accelerated metabolic demands and is associated with increased whole body substrate availability, including circulating lipids. The hypothesis that increases in hepatic glucagon action stimulate AMP-activated protein kinase (AMPK) signaling and peroxisome proliferator-activated receptor-α (PPARα) and fibroblast growth factor 21 (FGF21) expression in a manner modulated by fatty acids was tested in vivo. Wild-type (gcgr(+/+)) and glucagon receptor-null (gcgr(-/-)) littermate mice were studied using an 18-h fast, exercise, and hyperglucagonemic-euglycemic clamps plus or minus increased circulating lipids. Fasting and exercise in gcgr(+/+), but not gcgr(-/-) mice, increased hepatic phosphorylated AMPKα at threonine 172 (p-AMPK(Thr(172))) and PPARα and FGF21 mRNA. Clamp results in gcgr(+/+) mice demonstrate that hyperlipidemia does not independently impact or modify glucagon-stimulated increases in hepatic AMP/ATP, p-AMPK(Thr(172)), or PPARα and FGF21 mRNA. It blunted glucagon-stimulated acetyl-CoA carboxylase phosphorylation, a downstream target of AMPK, and accentuated PPARα and FGF21 expression. All effects were absent in gcgr(-/-) mice. These findings demonstrate that glucagon exerts a critical regulatory role in liver to stimulate pathways linked to lipid metabolism in vivo and shows for the first time that effects of glucagon on PPARα and FGF21 expression are amplified by a physiological increase in circulating lipids.


Asunto(s)
Adenilato Quinasa/metabolismo , Emulsiones Grasas Intravenosas/metabolismo , Factores de Crecimiento de Fibroblastos/biosíntesis , Glucagón/metabolismo , Hígado/metabolismo , PPAR alfa/biosíntesis , Adenilato Quinasa/genética , Animales , Área Bajo la Curva , Glucemia/metabolismo , Catecolaminas/sangre , Ácidos Grasos no Esterificados/sangre , Femenino , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Técnica de Clampeo de la Glucosa , Insulina/sangre , Hígado/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , PPAR alfa/genética , PPAR alfa/metabolismo , Condicionamiento Físico Animal/fisiología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores de Glucagón/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
4.
Eur J Cancer Prev ; 28(3): 188-195, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30640206

RESUMEN

Fish oil supplementation may represent a potential chemopreventive agent for reducing colorectal cancer risk. The mechanism of action of fish oil is unknown but presumed to be related to eicosanoid modification. The purpose of this study was to evaluate the effects of fish oil supplementation on the levels of urinary and rectal eicosanoids. We conducted a randomized, double-blind, controlled trial of 2.5 g of fish oil per day compared with olive oil supplementation over a 6-month period. Study participants had a history of colorectal adenomas. Randomization was stratified based on the gene variant rs174535 in the fatty acid desaturase 1 enzyme (FADS1), which affects tissue levels of arachidonic acid. A total of 141 participants were randomized. Urinary prostaglandin E2 metabolite (PGE-M) was measured at baseline, 3, and 6 months and rectal prostaglandin E2 (PGE2) at baseline and 6 months. Repeated-measures linear regression was used to determine the effect of the intervention on each outcome measure. Overall, fish oil supplementation was found to reduce urinary PGE-M production compared with olive oil (P=0.03). Fish oil did not reduce rectal PGE2 overall; however, it did significantly reduce PGE2 in the subgroup of participants not using aspirin or NSAIDs (P=0.04). FADS1 genotype did not seem to modify effects of fish oil on PGE2 production. We conclude that fish oil supplementation has a modest but beneficial effect on eicosanoids associated with colorectal carcinogenesis, particularly in those not taking aspirin or NSAIDs.


Asunto(s)
Adenoma/dietoterapia , Neoplasias Colorrectales/dietoterapia , Suplementos Dietéticos , Eicosanoides/metabolismo , Aceites de Pescado/administración & dosificación , Adenoma/etiología , Adenoma/metabolismo , Adenoma/patología , Anciano , Neoplasias Colorrectales/etiología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , delta-5 Desaturasa de Ácido Graso , Método Doble Ciego , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Factores de Riesgo
5.
Lipids ; 43(8): 703-11, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18481132

RESUMEN

Insulin resistance is characterized by increased metabolic uptake of fatty acids. Accordingly, techniques to examine in vivo shifts in fatty acid metabolism are of value in both clinical and experimental settings. Partially metabolizable long chain fatty acid (LCFA) tracers have been recently developed and employed for this purpose: [9,10-3H]-(R)-2-bromopalmitate ([3H]-BROMO) and [125I]-15-(rho-iodophenyl)-3-R,S-methylpentadecanoic acid ([125I]-BMIPP). These analogues are taken up like native fatty acids, but once inside the cell do not directly enter beta-oxidation. Rather, they become trapped in the slower processes of omega and alpha-oxidation. Study aims were to (1) simultaneously assess and compare [3H]-BROMO and [125I]-BMIPP and (2) determine if tracer breakdown is affected by elevated metabolic demands. Catheters were implanted in a carotid artery and jugular vein of Sprague-Dawley rats. Following 5 days recovery, fasted animals (5 h) underwent a rest (n = 8) or exercise (n = 8) (0.6 mi/h) protocol. An instantaneous bolus containing both [3H]-BROMO and [125I]-BMIPP was administered to determine LCFA uptake. No significant difference between [125I]-BMIPP and [3H]-BROMO uptake was found in cardiac or skeletal muscle during rest or exercise. In liver, rates of uptake were more than doubled with [3H]-BROMO compared to [125I]-BMIPP. Analysis of tracer conversion by TLC demonstrated no difference at rest. Exercise resulted in greater metabolism and excretion of tracers with approximately 37% and approximately 53% of [125I]-BMIPP and [3H]-BROMO present in conversion products at 40 min. In conclusion, [3H]-BROMO and [125I]-BMIPP are indistinguishable for the determination of tissue kinetics at rest in skeletal and cardiac muscle. Exercise preferentially exacerbates the breakdown of [3H]-BROMO, making [125I]-BMIPP the analogue of choice for prolonged (>30 min) experimental protocols with elevated metabolic demands.


Asunto(s)
Compuestos de Bromina/metabolismo , Ácidos Grasos/metabolismo , Yodobencenos/metabolismo , Palmitatos/metabolismo , Animales , Compuestos de Bromina/farmacocinética , Ácidos Grasos/farmacocinética , Radioisótopos de Yodo , Yodobencenos/farmacocinética , Masculino , Especificidad de Órganos , Palmitatos/farmacocinética , Ratas , Ratas Sprague-Dawley , Tritio
6.
JPEN J Parenter Enteral Nutr ; 31(4): 263-8, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17595432

RESUMEN

BACKGROUND: The chronic hemolytic anemia experienced by sickle cell disease (SCD) patients leads to adverse effects on oxygen transport by the blood and to a decrease in oxygen availability for peripheral tissues. Limited tissue oxygen availability has the potential to modify events of intracellular metabolism and, thus, alter lipid homeostasis. METHODS: The impact of SCD on plasma fatty acid homeostasis was determined in 8 African American SCD patients and in 6 healthy African American control subjects under postabsorptive conditions and during a 3-hour IV infusion of a nutrient solution containing lipid, glucose, and amino acids. RESULTS: SCD patients had higher fasting levels of plasma nonesterified fatty acids (NEFA), triglycerides, and phospholipids than healthy controls. Similarly, SCD patients had higher fasting levels of fatty acids in plasma triglycerides and phospholipids than healthy controls. Infusion of nutrients resulted in equivalent plasma NEFA profiles, total NEFA, and triglycerides in SCD patients and controls. However, the plasma phospholipid concentrations and fatty acid composition of plasma triglycerides and phospholipids were significantly higher in SCD patients; in particular, plasma pools of oleic acid were consistently increased in SCD. Plasma free oleic acid levels were elevated basally, leading to increased oleic acid content in triglycerides and phospholipids both post absorptively and during nutrient infusion. CONCLUSIONS: There is an underlying defect in lipid metabolism associated with SCD best manifested during the fasting state. This abnormality in lipid homeostasis has the potential to alter red blood cell (RBC) membrane fluidity and function in SCD patients.


Asunto(s)
Anemia de Células Falciformes/metabolismo , Membrana Eritrocítica/química , Metabolismo de los Lípidos , Adolescente , Adulto , Anemia de Células Falciformes/sangre , Estudios de Casos y Controles , Membrana Eritrocítica/metabolismo , Ayuno/sangre , Ayuno/metabolismo , Ácidos Grasos no Esterificados/sangre , Ácidos Grasos no Esterificados/química , Femenino , Humanos , Infusiones Parenterales , Metabolismo de los Lípidos/fisiología , Masculino , Persona de Mediana Edad , Fosfolípidos/sangre , Fosfolípidos/química , Periodo Posprandial/fisiología , Triglicéridos/sangre , Triglicéridos/química
7.
PLoS One ; 12(8): e0181046, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28793320

RESUMEN

Previous studies in our laboratory have established the presence of MTP in both white and brown adipose tissue in mice as well as in 3T3-L1 cells. Additional studies demonstrated an increase in MTP levels as 3T3-L1 cells differentiate into adipocytes concurrent with the movement of MTP from the juxtanuclear region of the cell to the surface of lipid droplets. This suggested a role for MTP in lipid droplet biogenesis and/or maturation. To probe the role of MTP in adipocytes, we used a Cre-Lox approach with aP2-Cre and Adipoq-Cre recombinase transgenic mice to knock down MTP expression in brown and white fat of mice. MTP expression was reduced approximately 55% in white fat and 65-80% in brown fat. Reducing MTP expression in adipose tissue had no effect on weight gain or body composition, whether the mice were fed a regular rodent or high fat diet. In addition, serum lipids and unesterified fatty acid levels were not altered in the knockdown mice. Importantly, decreased MTP expression in adipose tissue was associated with smaller lipid droplets in brown fat and smaller adipocytes in white fat. These results combined with our previous studies showing MTP lipid transfer activity is not necessary for lipid droplet initiation or growth in the early stages of differentiation, suggest that a structural feature of the MTP protein is important in lipid droplet maturation. We conclude that MTP protein plays a critical role in lipid droplet maturation, but does not regulate total body fat accumulation.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Proteínas Portadoras/metabolismo , Gotas Lipídicas/metabolismo , Células 3T3-L1 , Animales , Composición Corporal/genética , Proteínas Portadoras/genética , Dieta Alta en Grasa , Técnicas de Silenciamiento del Gen , Ratones , Ratones Transgénicos , Aumento de Peso/genética
8.
Diabetes ; 66(5): 1172-1184, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28246292

RESUMEN

Ten-week-old Zucker diabetic fatty (ZDF) rats at an early stage of diabetes embody metabolic characteristics of obese human patients with type 2 diabetes, such as severe insulin and glucose intolerance in muscle and the liver, excessive postprandial excursion of plasma glucose and insulin, and a loss of metabolic flexibility with decreased lipid oxidation. Metabolic flexibility and glucose flux were examined in ZDF rats during fasting and near-normal postprandial insulinemia and glycemia after correcting excessive postprandial hyperglycemia using treatment with a sodium-glucose cotransporter 2 inhibitor (SGLT2-I) for 7 days. Preprandial lipid oxidation was normalized, and with fasting, endogenous glucose production (EGP) increased by 30% and endogenous glucose disposal (E-Rd) decreased by 40%. During a postprandial hyperglycemic-hyperinsulinemic clamp after SGLT2-I treatment, E-Rd increased by normalizing glucose effectiveness to suppress EGP and stimulate hepatic glucose uptake; activation of glucokinase was restored and insulin action was improved, stimulating muscle glucose uptake in association with decreased intracellular triglyceride content. In conclusion, SGLT2-I treatment improves impaired glucose effectiveness in the liver and insulin sensitivity in muscle by eliminating glucotoxicity, which reinstates metabolic flexibility with restored preprandial lipid oxidation and postprandial glucose flux in ZDF rats.


Asunto(s)
Glucemia/efectos de los fármacos , Canagliflozina/farmacología , Intolerancia a la Glucosa/metabolismo , Hiperglucemia/metabolismo , Resistencia a la Insulina , Hígado/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Animales , Glucemia/metabolismo , Glucoquinasa/efectos de los fármacos , Glucoquinasa/metabolismo , Glucosa/metabolismo , Técnica de Clampeo de la Glucosa , Hipoglucemiantes , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/metabolismo , Músculo Esquelético/metabolismo , Oxidación-Reducción , Periodo Posprandial/efectos de los fármacos , Ratas , Ratas Zucker , Inhibidores del Cotransportador de Sodio-Glucosa 2
9.
Atherosclerosis ; 186(2): 267-74, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16144700

RESUMEN

Acyl-coenzyme A:cholesterol acyltransferase (ACAT) esterifies free cholesterol and stores cholesteryl esters in lipid droplets. Macrophage ACAT1 deficiency results in increased atherosclerotic lesion area in hyperlipidemic mice via disrupted cholesterol efflux, increased lipoprotein uptake, accumulation of intracellular vesicles, and accelerated apoptosis. The objective of this study was to determine whether lipid synthesis is affected by ACAT1. The synthesis, esterification, and efflux of new cholesterol were measured in peritoneal macrophages from ACAT1(-/-) mice. Cholesterol synthesis was increased by 134% (p=0.001) in ACAT1(-/-) macrophages compared to wildtype macrophages. Increased synthesis resulted in a proportional increase in the efflux of newly synthesized cholesterol. Although the esterification of new cholesterol was reduced by 93% (p<0.001) in ACAT1(-/-) macrophages, trace amounts of newly synthesized cholesteryl esters were detectable. Furthermore, the expression of SREBP1a mRNA was increased 6-fold in ACAT1(-/-) macrophages compared to wildtype macrophages, suggesting an up-regulation of cholesterol and fatty acid synthesis in ACAT1(-/-) macrophages. Increased cholesterol synthesis and up-regulation of SREBP in ACAT1(-/-) macrophages suggests that ACAT1 affects the regulation of lipid metabolism in macrophages. This change in cholesterol homeostasis may contribute to the atherogenic potential of ACAT1(-/-) macrophages.


Asunto(s)
Colesterol/biosíntesis , Macrófagos Peritoneales/enzimología , Esterol O-Aciltransferasa/deficiencia , Animales , Transporte Biológico , Células Cultivadas , Colesterol/sangre , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfolípidos/biosíntesis , Fosfolípidos/sangre , Esterol O-Aciltransferasa/fisiología
10.
Arterioscler Thromb Vasc Biol ; 25(1): 128-34, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15499044

RESUMEN

OBJECTIVE: Acyl-coenzyme A: cholesterol acyltransferase (ACAT) converts intracellular free cholesterol (FC) into cholesteryl esters (CE) for storage in lipid droplets. Recent studies in our laboratory have shown that the deletion of the macrophage ACAT1 gene results in apoptosis and increased atherosclerotic lesion area in the aortas of hyperlipidemic mice. The objective of the current study was to elucidate the mechanism of the increased atherosclerosis. METHODS AND RESULTS: CE storage and FC efflux were studied in ACAT1(-/-) peritoneal macrophages that were treated with acetylated low-density lipoprotein (acLDL). Our results show that efflux of cellular cholesterol was reduced by 25% in ACAT1-deficient cells compared with wild-type controls. This decrease occurred despite the upregulated expression of ABCA1, an important mediator of cholesterol efflux. In contrast, ACAT1 deficiency increased efflux of the cholesterol derived from acLDL by 32%. ACAT1-deficient macrophages also showed a 26% increase in the accumulation of FC derived from acLDL, which was associated with a 75% increase in the number of intracellular vesicles. CONCLUSIONS: Together, these data show that macrophage ACAT1 influences the efflux of both cellular and lipoprotein-derived cholesterol and propose a pathway for the pro-atherogenic transformation of ACAT1(-/-) macrophages.


Asunto(s)
Colesterol/metabolismo , Macrófagos Peritoneales/enzimología , Macrófagos Peritoneales/metabolismo , Esterol O-Aciltransferasa/deficiencia , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/biosíntesis , Transportadoras de Casetes de Unión a ATP/metabolismo , Acetil-CoA C-Acetiltransferasa , Animales , Transporte Biológico Activo/fisiología , Colesterol/toxicidad , Ésteres del Colesterol/metabolismo , LDL-Colesterol/química , LDL-Colesterol/metabolismo , Endosomas/química , Células Espumosas/metabolismo , Lisosomas/química , Macrófagos Peritoneales/química , Macrófagos Peritoneales/ultraestructura , Ratones , Microscopía Electrónica de Transmisión/métodos , Microscopía Fluorescente/métodos , ARN Mensajero/metabolismo , Esterol O-Aciltransferasa/fisiología , Tritio/metabolismo
11.
Sci Rep ; 6: 27308, 2016 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-27256115

RESUMEN

Microsomal triglyceride transfer protein (MTP) is a unique lipid transfer protein essential for the assembly of triglyceride-rich lipoproteins by the liver and intestine. Previous studies in mice identified a splice variant of MTP with an alternate first exon. Splice variants of human MTP have not been reported. Using PCR approaches we have identified two splice variants in human tissues, which we have named MTP-B and MTP-C. MTP-B has a unique first exon (Ex1B) located 10.5 kb upstream of the first exon (Ex1A) for canonical MTP (MTP-A); MTP-C contains both first exons for MTP-A and MTP-B. MTP-B was found in a number of tissues, whereas MTP-C was prominent in brain and testis. MTP-B does not encode a protein; MTP-C encodes the same protein encoded by MTP-A, although MTP-C translation is strongly inhibited by regulatory elements within its 5'-UTR. Using luciferase assays, we demonstrate that the promoter region upstream of exon 1B is quite adequate to drive expression of MTP. We conclude that alternate splicing plays a key role in regulating cellular MTP levels by introducing distinct promoter regions and unique 5'-UTRs, which contain elements that alter translation efficiency, enabling the cell to optimize MTP activity.


Asunto(s)
Empalme Alternativo , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/genética , Regulación de la Expresión Génica , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Humanos , Reacción en Cadena de la Polimerasa
12.
PLoS One ; 11(1): e0147252, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26771188

RESUMEN

Microsomal triglyceride transfer protein (MTP) is essential for the assembly of triglyceride-rich apolipoprotein B-containing lipoproteins. Previous studies in our laboratory identified a novel splice variant of MTP in mice that we named MTP-B. MTP-B has a unique first exon (1B) located 2.7 kB upstream of the first exon (1A) for canonical MTP (MTP-A). The two mature isoforms, though nearly identical in sequence and function, have different tissue expression patterns. In this study we report the identification of a second MTP splice variant (MTP-C), which contains both exons 1B and 1A. MTP-C is expressed in all the tissues we tested. In cells transfected with MTP-C, protein expression was less than 15% of that found when the cells were transfected with MTP-A or MTP-B. In silico analysis of the 5'-UTR of MTP-C revealed seven ATGs upstream of the start site for MTP-A, which is the only viable start site in frame with the main coding sequence. One of those ATGs was located in the 5'-UTR for MTP-A. We generated reporter constructs in which the 5'-UTRs of MTP-A or MTP-C were inserted between an SV40 promoter and the coding sequence of the luciferase gene and transfected these constructs into HEK 293 cells. Luciferase activity was significantly reduced by the MTP-C 5'-UTR, but not by the MTP-A 5'-UTR. We conclude that alternative splicing plays a key role in regulating MTP expression by introducing unique 5'-UTRs, which contain elements that alter translation efficiency, enabling the cell to optimize MTP levels and activity.


Asunto(s)
Proteínas Portadoras/metabolismo , Empalme Alternativo/genética , Animales , Células CHO , Proteínas Portadoras/genética , Cricetulus , Electroforesis en Gel de Poliacrilamida , Femenino , Células HEK293 , Humanos , Ratones , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN Mensajero/genética , Conejos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Diabetes ; 53(2): 336-46, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14747283

RESUMEN

Increased plasminogen activator inhibitor 1 (PAI-1) has been linked to not only thrombosis and fibrosis but also to obesity and insulin resistance. Increased PAI-1 levels have been presumed to be consequent to obesity. We investigated the interrelationships of PAI-1, obesity, and insulin resistance in a high-fat/high-carbohydrate (HF) diet-induced obesity model in wild-type (WT) and PAI-1-deficient mice (PAI-1(-/-)). Obesity and insulin resistance developing in WT mice on an HF diet were completely prevented in mice lacking PAI-1. PAI-1(-/-) mice on an HF diet had increased resting metabolic rates and total energy expenditure compared with WT mice, along with a marked increase in uncoupling protein 3 mRNA expression in skeletal muscle, likely mechanisms contributing to the prevention of obesity. In addition, insulin sensitivity was enhanced significantly in PAI-1(-/-) mice on an HF diet, as shown by euglycemic-hyperinsulinemic clamp studies. Peroxisome proliferator-activated receptor (PPAR)-gamma and adiponectin mRNA, key control molecules in lipid metabolism and insulin sensitivity, were maintained in response to an HF diet in white adipose tissue in PAI-1(-/-) mice, contrasting with downregulation in WT mice. This maintenance of PPAR-gamma and adiponectin may also contribute to the observed maintenance of body weight and insulin sensitivity in PAI-1(-/-) mice. Treatment in WT mice on an HF diet with the angiotensin type 1 receptor antagonist to downregulate PAI-1 indeed inhibited PAI-1 increases and ameliorated diet-induced obesity, hyperglycemia, and hyperinsulinemia. PAI-1 deficiency also enhanced basal and insulin-stimulated glucose uptake in adipose cells in vitro. Our data suggest that PAI-1 may not merely increase in response to obesity and insulin resistance, but may have a direct causal role in obesity and insulin resistance. Inhibition of PAI-1 might provide a novel anti-obesity and anti-insulin resistance treatment.


Asunto(s)
Resistencia a la Insulina/fisiología , Péptidos y Proteínas de Señalización Intercelular , Obesidad/prevención & control , Inhibidor 1 de Activador Plasminogénico/deficiencia , Adiponectina , Animales , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Calorimetría Indirecta , Proteínas Portadoras/genética , Modelos Animales de Enfermedad , Técnica de Clampeo de la Glucosa , Hiperinsulinismo , Insulina/administración & dosificación , Insulina/farmacología , Resistencia a la Insulina/genética , Canales Iónicos , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Proteínas Mitocondriales , Obesidad/sangre , Obesidad/genética , Inhibidor 1 de Activador Plasminogénico/genética , Inhibidor 1 de Activador Plasminogénico/fisiología , Reacción en Cadena de la Polimerasa , Proteínas/genética , ARN Mensajero/genética , Transcripción Genética , Triglicéridos/sangre , Triglicéridos/metabolismo , Proteína Desacopladora 1 , Aumento de Peso
14.
FEBS Lett ; 579(14): 3183-9, 2005 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-15922333

RESUMEN

Microsomal triglyceride transfer protein (MTP) is a carrier of triglyceride essential for the assembly of apolipoprotein (apo)B-containing lipoproteins by the liver and the small intestine. Its role in triglyceride transfer in tissues that do not secrete lipoproteins has not been explored. In particular, MTP would seem to be a candidate for a role in triglyceride metabolism within the adipocyte. To test this hypothesis, we probed adipocytes for the presence of MTP. Immunohistochemical and biochemical studies demonstrate MTP in adipocytes from brown and white fat depots of mice and human, as well as in 3T3-L1 cells. Confocal microscopy revealed MTP throughout 3T3 cells; however, MTP fluorescence was prominent in juxtanuclear areas. In differentiated 3T3 cells MTP fluorescence was very striking around lipid droplets. In vitro lipid transfer assays demonstrated the presence of triglyceride transfer activity within microsomal fractions isolated from rat adipose tissue. In addition, quantitative rtPCR studies showed that MTP expression in mouse white fat depots was approximately 1% of MTP expression in mouse liver. MTP mRNA in differentiated 3T3 cells was approximately 13% of liver expression. Our results provide unequivocal evidence for the presence of MTP in adipocytes and present new possibilities for defining the mechanisms by which triglyceride is stored and/or hydrolyzed and mobilized.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Proteínas Portadoras/metabolismo , Adipocitos/citología , Animales , Proteínas Portadoras/genética , Línea Celular , Grasas/metabolismo , Inmunohistoquímica , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , Triglicéridos/metabolismo
15.
Metabolism ; 54(10): 1309-15, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16154429

RESUMEN

Studies in our laboratory have shown that a fraction of apolipoprotein (apo) E internalized by hepatocytes escapes degradation and is resecreted. Although the intracellular routing is not fully understood, our studies suggest that a portion of apoE recycles through the Golgi apparatus. Given the role of the Golgi apparatus in lipoprotein secretion and the fact that apoE modulates the hepatic secretion of very low-density lipoprotein, we hypothesized that recycling apoE has an effect on hepatic very low-density lipoprotein assembly and/or secretion. To test this hypothesis, apoE-/- mice were transplanted with bone marrow from wild-type mice. In this model, extrahepatic (macrophage-derived) apoE is internalized by the hepatocytes in vivo and is resecreted when the hepatocytes are placed in culture. Unexpectedly, our studies demonstrate that recycling apoE has little effect on hepatic lipid content or hepatocyte triglyceride secretion. In addition, recycling apoE has little effect on the expression of enzymes and proteins involved in lipid synthesis as well as plasma lipoprotein apoproteins. We conclude that the physiological relevance of apoE recycling may not be related to cell-specific functions, such as lipoprotein assembly in the liver. Rather, recycling may provide a mechanism for modulating general cellular effects such as intracellular cholesterol transport or cholesterol efflux.


Asunto(s)
Apolipoproteínas E/metabolismo , Hepatocitos/metabolismo , Animales , Trasplante de Médula Ósea , Proteínas Potenciadoras de Unión a CCAAT/genética , Células Cultivadas , Proteínas de Unión al ADN/genética , Ácidos Grasos/análisis , Lipoproteínas VLDL/metabolismo , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/análisis , Proteína 1 de Unión a los Elementos Reguladores de Esteroles , Factores de Transcripción/genética , Triglicéridos/análisis
16.
Arterioscler Thromb Vasc Biol ; 24(11): 2143-8, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15374850

RESUMEN

OBJECTIVE: We evaluated the role of angiotensin II (AII) in a marrow-derived macrophage-driven model of atherosclerosis. METHODS AND RESULTS: Eight-week-old C57BL/6 wild-type mice were reconstituted with bone marrow harvested from apolipoprotein E-deficient (apoE-/---> apoE+/+) or wild-type for apoE gene (apoE+/+--> apoE+/+) mice. At 20 weeks, mice were exposed to either AII (1000 ng/kg per minute subcutaneously) or saline for 2 weeks. Animals did not differ in body weight, blood pressure, cholesterol/triglycerides, or peripheral blood monocyte counts. ApoE-/---> apoE+/+ mice exposed to AII had 3-fold greater atherosclerotic area than saline-treated apoE-/---> apoE+/+ mice. By contrast, AII did not affect atherosclerosis in apoE+/+--> apoE+/+ mice. Macrophage-positive areas were increased by AII in mice reconstituted with either apoE-deficient or apoE-competent marrow. AII also significantly increased fragmentation of elastin laminae in both apoE-/---> apoE+/+ and apoE+/+--> apoE+/+ mice. In vitro, AII caused greater increase in monocyte chemoattractant protein-1-stimulated migration of macrophages harvested from AII-infused versus saline-infused mice. CONCLUSIONS: The current studies reveal that AII has both initiating and sustaining proatherogenic effects. By promoting macrophage migration into the vascular intima, AII is pivotal in initiating atherosclerosis; by promoting elastin breaks, a novel mechanism implicated in migration and proliferation of smooth muscle cells, AII may be pivotal in subsequent development and expansion of atherosclerotic lesion.


Asunto(s)
Angiotensina II/farmacología , Arteriosclerosis/metabolismo , Arteriosclerosis/patología , Macrófagos/metabolismo , Angiotensina II/administración & dosificación , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/fisiología , Trasplante de Médula Ósea/métodos , Movimiento Celular/efectos de los fármacos , Colesterol/metabolismo , Modelos Animales de Enfermedad , Femenino , Bombas de Infusión Implantables , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos C57BL
17.
PLoS One ; 10(8): e0135598, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26267806

RESUMEN

Lipid droplets are intracellular energy storage organelles composed of a hydrophobic core of neutral lipid, surrounded by a monolayer of phospholipid and a diverse array of proteins. The function of the vast majority of these proteins with regard to the formation and/or turnover of lipid droplets is unknown. Our laboratory was the first to report that microsomal triglyceride transfer protein (MTP), a lipid transfer protein essential for the assembly of triglyceride-rich lipoproteins, was expressed in adipose tissue of humans and mice. In addition, our studies suggested that MTP was associated with lipid droplets in both brown and white fat. Our observations led us to hypothesize that MTP plays a key role in lipid droplet formation and/or turnover. The objective of these studies was to gain insight into the function of MTP in adipocytes. Using molecular, biochemical, and morphologic approaches we have shown: 1) MTP protein levels increase nearly five-fold as 3T3-L1 cells differentiate into adipocytes. 2) As 3T3-L1 cells undergo differentiation, MTP moves from the juxtanuclear region of the cell to the surface of lipid droplets. MTP and perilipin 2, a major lipid droplet surface protein, are found on the same droplets; however, MTP does not co-localize with perilipin 2. 3) Inhibition of MTP activity has no effect on the movement of triglyceride out of the cell either as a lipid complex or via lipolysis. 4) MTP is found associated with lipid droplets within hepatocytes from human fatty livers, suggesting that association of MTP with lipid droplets is not restricted to adipocytes. In summary, our data demonstrate that MTP is a lipid droplet-associated protein. Its location on the surface of the droplet in adipocytes and hepatocytes, coupled with its known function as a lipid transfer protein and its increased expression during adipocyte differentiation suggest a role in lipid droplet biology.


Asunto(s)
Adipocitos/metabolismo , Proteínas Portadoras/metabolismo , Citosol/metabolismo , Gotas Lipídicas/metabolismo , Células 3T3-L1 , Animales , Proteínas Portadoras/genética , Diferenciación Celular/fisiología , Electroforesis en Gel de Poliacrilamida , Inmunohistoquímica , Ratones , Microscopía Fluorescente , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
J Am Assoc Lab Anim Sci ; 54(4): 378-83, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26224437

RESUMEN

Sexually mature zebrafish were housed as single male-female pairs with or without plastic vegetation for 1, 5, or 10 d for comparison of whole-body cortisol measured by radioimmunoassay. Individually housed male zebrafish were used as controls. In the fish that were pair-housed without vegetation (NVeg), one animal died in 5 of 24 pairs, and one animal was alive but wounded in an additional pair. No deaths or wounds occurred in the fish that were pair-housed with vegetation (Veg). Cortisol levels did not differ between the treatment groups on day 1. On day 5, cortisol values were higher in the Veg group than in the individually housed fish (P < 0.0005) and the NVeg fish (P = 0.004). On day 10, the relationships were inversed: cortisol levels had risen in the individually housed and NVeg groups and had fallen to baseline levels in the Veg group. Cortisol values on day 10 were lower in the Veg group than in the individually housed (P = 0.004) and NVeg (P = 0.05) groups. Cortisol levels in individually housed male zebrafish increased over time. Although this study did not demonstrate a reduction in cortisol levels associated with providing vegetation, this enrichment prevented injury and death from fighting. These findings show how commonly used housing situations may affect the wellbeing of laboratory zebrafish.


Asunto(s)
Vivienda para Animales , Pez Cebra , Agresión , Animales , Ecosistema , Femenino , Hidrocortisona/análisis , Masculino
19.
Free Radic Biol Med ; 34(12): 1631-5, 2003 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-12788483

RESUMEN

While studies showed that aging is accompanied by increased exposure of the brain to oxidative stress, others have not detected any age-correlated differences in levels of markers of oxidative stress. Use of conventional markers of oxidative damage in vivo, which may be formed ex vivo and/or eliminated by endogenous metabolism, may explain these conflicting results. Recently, F(2)-isoprostanes and F(4)-neuroprostanes, peroxidation products of arachidonic acid and docosahexaenoic acid, respectively, have been identified as sensitive and reliable markers of oxidative injury. Therefore, this study was designed to quantify brain levels of F(2)-isoprostanes and F(4)-neuroprostanes and their precursors in 4, 10, 50, and 100 week old male Fischer 344 rats. Data show that levels of F(2)-isoprostanes and F(4)-neuroprostanes were comparable in all animal age groups. However, levels of F(4)-neuroprostanes were approximately 20-fold higher than those of F(2)-isoprostanes in all age groups, despite the fact that brain levels of docosahexaenoic acid were only twice as high as those of arachidonic acid. Based on our findings, it is concluded that aging is not accompanied by enhanced brain susceptibility to oxidative stress. Furthermore, the metabolically active gray matter of the brain, where docosahexaenoic acid is abundant, appears more susceptible to oxidative stress than the white matter.


Asunto(s)
Envejecimiento/fisiología , Encéfalo/metabolismo , Dinoprostona/metabolismo , F2-Isoprostanos/metabolismo , Estrés Oxidativo , Distribución por Edad , Animales , Ácido Araquidónico/metabolismo , Encéfalo/patología , Ácidos Docosahexaenoicos/metabolismo , Masculino , Ratas , Ratas Endogámicas F344
20.
J Alzheimers Dis ; 4(1): 19-30, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12214015

RESUMEN

Recent studies of cerebrospinal fluid (CSF) have shown increased oxidation of CSF lipoproteins in Alzheimer's disease (AD) patients, and neurotoxicity from oxidized CSF lipoproteins in culture. Since inheritance of different alleles of the apolipoprotein (apo) E gene is a risk factor for AD and apoE is the major lipoprotein trafficking molecule in brain, we hypothesized that apoE may modify the pathogenesis of AD by directing the delivery of oxidized CSF lipoproteins to neurons. To test this hypothesis, we adapted a method previously used with isolated plasma lipoproteins to specifically label lipid particles in situ in native CSF and quantified their delivery to human SK-N-BE(2)C neuroblastoma cells. CSF lipoproteins were delivered to neuronal cells largely through apoE-dependent processes. Importantly, CSF lipoproteins from AD patients were delivered more efficiently than CSF lipoproteins from age-matched controls; this effect was not associated with apoE genotype or degree of CSF lipoprotein oxidation but was associated with apoE monomer concentration that tended to be lower in AD patients. The inverse relationship between apoE monomer concentration and CSF lipoprotein delivery was duplicated in SK-N-BE(2)C cells, but not human astrocytoma cells, using artificial lipid particles and purified human apoE. These results suggest that lipoproteins in CSF from AD patients are delivered more efficiently to neurons than are CSF lipoproteins from controls, and that this abnormality may be explained largely by variations in CSF apoE concentration.


Asunto(s)
Enfermedad de Alzheimer/líquido cefalorraquídeo , Apolipoproteínas E/fisiología , Peroxidación de Lípido/fisiología , Lipoproteínas/líquido cefalorraquídeo , Alelos , Enfermedad de Alzheimer/genética , Apolipoproteína E3 , Apolipoproteína E4 , Apolipoproteínas E/genética , Encéfalo/metabolismo , Colesterol/líquido cefalorraquídeo , Humanos , Peroxidación de Lípido/genética , Neuroblastoma , Neuronas/metabolismo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA