Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Biol Chem ; 296: 100344, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33524391

RESUMEN

A low-sodium (LS) diet has been shown to reduce blood pressure (BP) and the incidence of cardiovascular diseases. However, severe dietary sodium restriction promotes insulin resistance (IR) and dyslipidemia in animal models and humans. Thus, further clarification of the long-term consequences of LS is needed. Here, we investigated the effects of chronic LS on gastrocnemius gene and protein expression and lipidomics and its association with IR and plasma lipids in LDL receptor knockout mice. Three-month-old male mice were fed a normal sodium diet (NS; 0.5% Na; n = 12-19) or LS (0.06% Na; n = 14-20) over 90 days. Body mass (BM), BP, plasma total cholesterol, triacylglycerol (TG), glucose, hematocrit, and IR were evaluated. LS increased BM (9%), plasma TG (51%), blood glucose (19%), and IR (46%) when compared with the NS. RT-qPCR analysis revealed that genes involved in lipid uptake and oxidation were increased by the LS: Fabp3 (106%), Prkaa1 (46%), and Cpt1 (74%). Genes and proteins (assessed by Western blotting) involved in insulin signaling were not changed by the LS. Similarly, lipid species classically involved in muscle IR, such as diacylglycerols and ceramides detected by ultra-high-performance liquid chromatography coupled to mass spectrometry, were also unchanged by LS. Species of phosphatidylcholines (68%), phosphatidylinositol (90%), and free fatty acids (59%) increased while cardiolipins (41%) and acylcarnitines (9%) decreased in gastrocnemius in response to LS and were associated with glucose disposal rate. Together these results suggest that chronic LS alters glycerophospholipid and fatty acids species in gastrocnemius that may contribute to glucose and lipid homeostasis derangements in mice.


Asunto(s)
Dieta Hiposódica , Resistencia a la Insulina , Metabolismo de los Lípidos , Músculo Esquelético/metabolismo , Animales , Lipidómica , Masculino , Ratones , Sodio en la Dieta/metabolismo
2.
Cardiovasc Diabetol ; 11: 100, 2012 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-22897936

RESUMEN

BACKGROUND: Metabolic syndrome is characterized by insulin resistance, which is closely related to GLUT4 content in insulin-sensitive tissues. Thus, we evaluated the GLUT4 expression, insulin resistance and inflammation, characteristics of the metabolic syndrome, in an experimental model. METHODS: Spontaneously hypertensive neonate rats (18/group) were treated with monosodium glutamate (MetS) during 9 days, and compared with Wistar-Kyoto (C) and saline-treated SHR (H). Blood pressure (BP) and lipid levels, C-reactive protein (CRP), interleukin 6 (IL-6), TNF-α and adiponectin were evaluated. GLUT4 protein was analysed in the heart, white adipose tissue and gastrocnemius. Studies were performed at 3 (3-mo), 6 (6-mo) and 9 (9-mo) months of age. RESULTS: MetS rats were more insulin resistant (p<0.001, all ages) and had higher BP (3-mo: p<0.001, 6-mo: p = 0.001, 9-mo: p = 0.015) as compared to C. At 6 months, CRP, IL-6 and TNF-α were higher (p<0.001, all comparisons) in MetS rats vs H, but adiponectin was lower in MetS at 9 months (MetS: 32 ± 2, H: 42 ± 2, C: 45 ± 2 pg/mL; p<0.001). GLUT4 protein was reduced in MetS as compared to C rats at 3, 6 and 9-mo, respectively (Heart: 54%, 50% and 57%; Gastrocnemius: 37%, 56% and 50%; Adipose tissue: 69%, 61% and 69%). CONCLUSIONS: MSG-treated SHR presented all metabolic syndrome characteristics, as well as reduced GLUT4 content, which must play a key role in the impaired glycemic homeostasis of the metabolic syndrome.


Asunto(s)
Transportador de Glucosa de Tipo 4/metabolismo , Mediadores de Inflamación/sangre , Resistencia a la Insulina , Síndrome Metabólico/metabolismo , Adiponectina/sangre , Tejido Adiposo Blanco/metabolismo , Animales , Animales Recién Nacidos , Biomarcadores/sangre , Presión Sanguínea , Proteína C-Reactiva/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Hipertensión/sangre , Hipertensión/complicaciones , Hipertensión/fisiopatología , Interleucina-6/sangre , Lípidos/sangre , Masculino , Síndrome Metabólico/sangre , Síndrome Metabólico/inducido químicamente , Síndrome Metabólico/inmunología , Síndrome Metabólico/fisiopatología , Músculo Esquelético/metabolismo , Miocardio/metabolismo , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Glutamato de Sodio , Factores de Tiempo , Factor de Necrosis Tumoral alfa/sangre
3.
Am J Physiol Endocrinol Metab ; 299(6): E1028-37, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20841505

RESUMEN

Salivary gland dysfunction is a feature in diabetes and hypertension. We hypothesized that sodium-glucose cotransporter 1 (SGLT1) participates in salivary dysfunctions through a sympathetic- and protein kinase A (PKA)-mediated pathway. In Wistar-Kyoto (WKY), diabetic WKY (WKY-D), spontaneously hypertensive (SHR), and diabetic SHR (SHR-D) rats, PKA/SGLT1 proteins were analyzed in parotid and submandibular glands, and the sympathetic nerve activity (SNA) to the glands was monitored. Basal SNA was threefold higher in SHR (P < 0.001 vs. WKY), and diabetes decreased this activity (∼50%, P < 0.05) in both WKY and SHR. The catalytic subunit of PKA and the plasma membrane SGLT1 content in acinar cells were regulated in parallel to the SNA. Electrical stimulation of the sympathetic branch to salivary glands increased (∼30%, P < 0.05) PKA and SGLT1 expression. Immunohistochemical analysis confirmed the observed regulations of SGLT1, revealing its location in basolateral membrane of acinar cells. Taken together, our results show highly coordinated regulation of sympathetic activity upon PKA activity and plasma membrane SGLT1 content in salivary glands. Furthermore, the present findings show that diabetic- and/or hypertensive-induced changes in the sympathetic activity correlate with changes in SGLT1 expression in basolateral membrane of acinar cells, which can participate in the salivary glands dysfunctions reported by patients with these pathologies.


Asunto(s)
Membrana Celular/metabolismo , Diabetes Mellitus/metabolismo , Hipertensión/metabolismo , Glándula Parótida/metabolismo , Transportador 1 de Sodio-Glucosa/metabolismo , Glándula Submandibular/metabolismo , Sistema Nervioso Simpático/metabolismo , Análisis de Varianza , Animales , Western Blotting , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Diabetes Mellitus/fisiopatología , Hipertensión/fisiopatología , Inmunohistoquímica , Masculino , Glándula Parótida/fisiopatología , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Transducción de Señal , Glándula Submandibular/fisiopatología , Sistema Nervioso Simpático/fisiopatología
4.
Mol Cell Endocrinol ; 481: 26-34, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30528377

RESUMEN

Expression of the glucose transporter GLUT4, encoded by Slc2a4 gene, is reduced in both type 1 and type 2 diabetes (T1D and T2D), contributing to glycemic impairment. The present study investigated epigenetic regulations at the Slc2a4 promoter in skeletal muscle of T1D- and T2D-like experimental models. Slc2a4/GLUT4 repression was observed in T1D and T2D and that was reversed by insulin and resveratrol treatments, respectively. In both T1D-like and T2D-like animals, tri-methylation at lysine 9 of histone 3 (H3K9me3) increased in the Slc2a4 enhancer segment, whereas MEF2A/D binding into this segment was reduced; all effects were reversed by respective treatments. This study reveals that increased H3K9me3 in the Slc2a4 promoter enhancer segment contributes to reduce GLUT4 expression in skeletal muscle and to worse glycemic control in diabetes, pointing to the H3K9me3 of Slc2a4 promoter as a potential target for development of new approaches for treating diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Transportador de Glucosa de Tipo 4/genética , Histonas/metabolismo , Músculo Esquelético/metabolismo , Animales , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 2/genética , Modelos Animales de Enfermedad , Epigénesis Genética , Transportador de Glucosa de Tipo 4/metabolismo , Histonas/química , Humanos , Insulina , Lisina/metabolismo , Masculino , Metilación , Ratones , Regiones Promotoras Genéticas , Ratas , Resveratrol
5.
Sci Rep ; 8(1): 8109, 2018 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-29802324

RESUMEN

Little is known about advanced glycation end products (AGEs) participation in glucose homeostasis, a process in which skeletal muscle glucose transporter GLUT4 (Scl2a4 gene) plays a key role. This study investigated (1) the in vivo and in vitro effects of AGEs on Slc2a4/GLUT4 expression in skeletal muscle of healthy rats, and (2) the potential involvement of endoplasmic reticulum and inflammatory stress in the observed regulations. For in vivo analysis, rats were treated with advanced glycated rat albumin (AGE-albumin) for 12 weeks; for in vitro analysis, soleus muscles from normal rats were incubated with bovine AGE-albumin for 2.5 to 7.5 hours. In vivo, AGE-albumin induced whole-body insulin resistance; decreased (~30%) Slc2a4 mRNA and GLUT4 protein content; and increased (~30%) the nuclear content of nuclear factor NF-kappa-B p50 subunit (NFKB1), and cellular content of 78 kDa glucose-regulated protein (GRP78). In vitro, incubation with AGE-albumin decreased (~50%) the Slc2a4/GLUT4 content; and increased cellular content of GRP78/94, phosphorylated-IKK-alpha/beta, nuclear content of NFKB1 and RELA, and the nuclear protein binding into Slc2a4 promoter NFKB-binding site. The data reveal that AGEs impair glucose homeostasis in non-diabetic states of increased AGEs concentration; an effect that involves activation of endoplasmic reticulum- and inflammatory-stress and repression of Slc2a4/GLUT4 expression.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Transportador de Glucosa de Tipo 4/genética , Productos Finales de Glicación Avanzada/farmacología , Resistencia a la Insulina , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Animales , Biomarcadores/metabolismo , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Glucosa/metabolismo , Homeostasis/efectos de los fármacos , Masculino , FN-kappa B/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar
6.
Mol Cell Endocrinol ; 447: 116-124, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28238722

RESUMEN

Because of the paucity of information regarding metabolic effects of advanced glycation end products (AGEs) on liver, we evaluated effects of AGEs chronic administration in (1) insulin sensitivity; (2) hepatic expression of genes involved in AGEs, glucose and fat metabolism, oxidative stress and inflammation and; (3) hepatic morphology and glycogen content. Rats received intraperitoneally albumin modified (AlbAGE) or not by advanced glycation for 12 weeks. AlbAGE induced whole-body insulin resistance concomitantly with increased hepatic insulin sensitivity, evidenced by activation of AKT, inactivation of GSK3, increased hepatic glycogen content, and decreased expression of gluconeogenesis genes. Additionally there was reduction in hepatic fat content, in expression of lipogenic, pro-inflamatory and pro-oxidative genes and increase in reactive oxygen species and in nuclear expression of NRF2, a transcription factor essential to cytoprotective response. Although considered toxic, AGEs become protective when administered chronically, stimulating AKT signaling, which is involved in cellular defense and insulin sensitivity.


Asunto(s)
Productos Finales de Glicación Avanzada/farmacología , Hormesis/efectos de los fármacos , Resistencia a la Insulina , Hígado/metabolismo , Albúminas/farmacología , Animales , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Gluconeogénesis/efectos de los fármacos , Gluconeogénesis/genética , Productos Finales de Glicación Avanzada/administración & dosificación , Glucógeno/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Proteína HMGB1/metabolismo , Mediadores de Inflamación/metabolismo , Inyecciones Intraperitoneales , Lipogénesis/efectos de los fármacos , Lipogénesis/genética , Hígado/efectos de los fármacos , Masculino , Modelos Biológicos , Factor 2 Relacionado con NF-E2/metabolismo , Oxidación-Reducción , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo
7.
Am J Med Sci ; 331(6): 309-14, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16775437

RESUMEN

BACKGROUND: Salt restriction is recommended for hypertension treatment to reduce blood pressure, but its effect on some risk factors is still a matter of discussion. The aim of this study was to observe the effect of a long period of salt restriction or overload on blood pressure, left ventricular mass (LVM), kidney mass (KM), glucose tolerance, and plasma insulin. METHODS: Male Wistar rats were fed from weaning with a low-salt diet (LSD) or a high-salt diet (HSD) until 72 weeks of age. After 48 weeks, the diets were changed in half of the rats: HSD until 48 weeks and then LSD (LHSD) and LSD until 48 weeks and then HSD (HLSD). Body weight, blood pressure, electrolyte excretion, creatinine clearance, plasma renin activity, LVM, KM, and intravenous glucose tolerance test with insulin determinations were evaluated. RESULTS: Blood pressure, LVM and KM were higher on the HSD than on the LSD. Blood pressure was lower on the LHSD than on the HLSD. There were no differences in LVM and KM on the LHSD compared with the HLSD. The relationship between area under the curve (AUC) of insulin and glucose during the intravenous glucose tolerance test was higher on the LSD. No differences were detected in AUC between the two groups of rats whose diet were inverted with 48 weeks of age. CONCLUSIONS: A chronic HSD increases blood pressure, LVM, and KM and a chronic LSD increases plasma insulin in response to a glucose challenge in aging rats. The hypotensive effect of salt restriction is not modified by a previous long period on a HSD.


Asunto(s)
Presión Sanguínea/efectos de los fármacos , Dieta Hiposódica , Corazón/efectos de los fármacos , Insulina/sangre , Cloruro de Sodio Dietético/administración & dosificación , Cloruro de Sodio Dietético/farmacología , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Área Bajo la Curva , Peso Corporal , Prueba de Tolerancia a la Glucosa , Ventrículos Cardíacos/efectos de los fármacos , Resistencia a la Insulina , Riñón/efectos de los fármacos , Masculino , Miocardio/patología , Ratas , Ratas Wistar , Renina/sangre
8.
Life Sci ; 93(22): 805-13, 2013 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-24157454

RESUMEN

AIMS: Solute carrier 2a2 (Slc2a2) gene codifies the glucose transporter GLUT2, a key protein for glucose flux in hepatocytes and renal epithelial cells of proximal tubule. In diabetes mellitus, hepatic and tubular glucose output has been related to Slc2a2/GLUT2 overexpression; and controlling the expression of this gene may be an important adjuvant way to improve glycemic homeostasis. Thus, the present study investigated transcriptional mechanisms involved in the diabetes-induced overexpression of the Slc2a2 gene. MAIN METHODS: Hepatocyte nuclear factors 1α and 4α (HNF-1α and HNF-4α), forkhead box A2 (FOXA2), sterol regulatory element binding protein-1c (SREBP-1c) and the CCAAT-enhancer-binding protein (C/EBPß) mRNA expression (RT-PCR) and binding activity into the Slc2a2 promoter (electrophoretic mobility assay) were analyzed in the liver and kidney of diabetic and 6-day insulin-treated diabetic rats. KEY FINDINGS: Slc2a2/GLUT2 expression increased by more than 50% (P<0.001) in the liver and kidney of diabetic rats, and 6-day insulin treatment restores these values to those observed in non-diabetic animals. Similarly, the mRNA expression and the binding activity of HNF-1α, HNF-4α and FOXA2 increased by 50 to 100% (P<0.05 to P<0.001), also returning to values of non-diabetic rats after insulin treatment. Neither the Srebf1 and Cebpb mRNA expression, nor the SREBP-1c and C/EBP-ß binding activity was altered in diabetic rats. SIGNIFICANCE: HNF-1α, HNF-4α and FOXA2 transcriptional factors are involved in diabetes-induced overexpression of Slc2a2 gene in the liver and kidney. These data point out that these transcriptional factors are important targets to control GLUT2 expression in these tissues, which can contribute to glycemic homeostasis in diabetes.


Asunto(s)
Diabetes Mellitus Experimental/genética , Transportador de Glucosa de Tipo 2/genética , Factor Nuclear 1-alfa del Hepatocito/genética , Factor Nuclear 3-beta del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/genética , Riñón/metabolismo , Hígado/metabolismo , Animales , Sitios de Unión , Proteína beta Potenciadora de Unión a CCAAT/genética , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Transportador de Glucosa de Tipo 2/metabolismo , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Factor Nuclear 3-beta del Hepatocito/metabolismo , Factor Nuclear 4 del Hepatocito/metabolismo , Insulina/farmacología , Riñón/efectos de los fármacos , Hígado/efectos de los fármacos , Masculino , Regiones Promotoras Genéticas , Ratas , Ratas Wistar , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética
9.
Eur J Pharmacol ; 689(1-3): 285-93, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22713545

RESUMEN

Quercetin is a potent anti-inflammatory flavonoid, but its capacity to modulate insulin sensitivity in obese insulin resistant conditions is unknown. This study investigated the effect of quercetin treatment upon insulin sensitivity of ob/ob mice and its potential molecular mechanisms. Obese ob/ob mice were treated with quercetin for 10 weeks, and L6 myotubes were treated with either palmitate or tumor necrosis factor-α (TNFα) plus quercetin. Cells and muscles were processed for analysis of glucose transporter 4 (GLUT4), TNFα and inducible nitric oxide synthase (iNOS) expression, and c-Jun N-terminal kinase (JNK) and inhibitor of nuclear factor-κB (NF-κB) kinase (IκK) phosphorylation. Myotubes were assayed for glucose uptake and NF-κB translocation. Chromatin immunoprecipitation assessed NF-κB binding to GLUT4 promoter. Quercetin treatment improved whole body insulin sensitivity by increasing GLUT4 expression and decreasing JNK phosphorylation, and TNFα and iNOS expression in skeletal muscle. Quercetin suppressed palmitate-induced upregulation of TNFα and iNOS and restored normal levels of GLUT4 in myotubes. In parallel, quercetin suppressed TNFα-induced reduction of glucose uptake in myotubes. Nuclear accumulation of NF-κB in myotubes and binding of NF-κB to GLUT4 promoter in muscles of ob/ob mice were also reduced by quercetin. We demonstrated that quercetin decreased the inflammatory status in skeletal muscle of obese mice and in L6 myotubes. This effect was followed by increased muscle GLUT4, with parallel improvement of insulin sensitivity. These results point out quercetin as a putative strategy to manage inflammatory-related insulin resistance.


Asunto(s)
Mediadores de Inflamación/antagonistas & inhibidores , Insulina/fisiología , Fibras Musculares Esqueléticas/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Quercetina/farmacología , Animales , Antioxidantes/farmacología , Regulación hacia Abajo/efectos de los fármacos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/prevención & control , Mediadores de Inflamación/metabolismo , Masculino , Ratones , Ratones Obesos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Quercetina/uso terapéutico , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA