Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Fish Physiol Biochem ; 46(4): 1207-1218, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32212006

RESUMEN

Glucose transporter 2 (glut2) has been studied in mammals, aves, and several fish, while the comparative studies of glut2 in common carp are still lacking. In this study, glut2 was firstly isolated and characterized from the liver of common carp. The full-length cDNA of glut2 was 2351 bp with an open reading frame (ORF) of 1512 bp, encoding 503 amino acids. Alignment of glut2 amino acid sequences from different species revealed that common carp glut2 showed higher sequence identity with teleosts, and lower homology with mammals and amphibians. Tissue distribution demonstrated that glut2 mRNA level was mainly expressed in liver, foregut, and midgut. To investigate the actions of glut2 on glucose metabolism, the level of glut2 mRNA was detected after intraperitoneal injection of glucose, human insulin and glucagon (100 ng/g), respectively. Following glucose administration, glut2 gene expression was significantly upregulated at 3 h in the foregut. However, no change was found in hepatic glut2 mRNA level, indicating that glut2 may have a role in intestinal glucose uptake rather than in the liver. Following insulin treatment, the expression of glut2 was markedly downregulated at 3 h and 6 h in the liver, and at 3 h in the foregut, respectively. Furthermore, glut2 mRNA expression was unaffected by glucagon injection in the liver and foregut. These results suggested that the expression of glut2 regulated by pancreatic hormones was different. Taken together, our studies firstly revealed the structure of the glut2 gene and its potential functions in glucose metabolism of common carp.


Asunto(s)
Carpas/metabolismo , Transportador de Glucosa de Tipo 2/genética , Secuencia de Aminoácidos , Aminoácidos/genética , Análisis de Varianza , Animales , Secuencia de Bases , Clonación Molecular , Regulación hacia Abajo , Expresión Génica , Glucagón/administración & dosificación , Glucagón/farmacología , Glucosa/administración & dosificación , Glucosa/farmacología , Transportador de Glucosa de Tipo 2/efectos de los fármacos , Transportador de Glucosa de Tipo 2/aislamiento & purificación , Transportador de Glucosa de Tipo 2/metabolismo , Insulina/administración & dosificación , Insulina/farmacología , Mucosa Intestinal/metabolismo , Hígado/química , Hígado/metabolismo , Sistemas de Lectura Abierta/genética , Filogenia , Distribución Aleatoria , Reacción en Cadena en Tiempo Real de la Polimerasa , Alineación de Secuencia , Regulación hacia Arriba
2.
Endocr J ; 67(1): 73-80, 2020 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-31611477

RESUMEN

Those who smoke nicotine-based cigarettes have elevated plasma levels of ghrelin, a hormone secreted from the stomach. Ghrelin has various physiological functions and has recently been shown to be involved in regulating biological rhythms. Therefore, in this study, in order to clarify the significance of the plasma ghrelin increase in smokers, we sought to clarify how nicotine and ghrelin affect the expression dynamics of clock genes using a mouse model. A single dose of nicotine administered intraperitoneally increased plasma ghrelin concentrations transiently, whereas continuous administration of nicotine with an osmotic minipump did not induce any change in the plasma ghrelin concentration. Single administration of nicotine resulted in a transient increase in ghrelin gene expression in the pancreas but not in the stomach, which is the major producer of ghrelin. In addition, in the pancreas, the expression of clock genes was also increased temporarily. Therefore, in order to clarify the interaction between nicotine-induced ghrelin gene expression and clock gene expression in the pancreas, nicotine was administered to ghrelin gene-deficient mice. Administration of nicotine to ghrelin-gene deficient mice increased clock gene expression in the pancreas. However, upon nicotine administration to mice pretreated with octanoate to upregulate ghrelin activity, expression levels of nicotine-inducible clock genes in the pancreas were virtually the same as those in mice not administered nicotine. Thus, our findings indicate that pancreatic ghrelin may suppress nicotine-induced clock gene expression in the pancreas.


Asunto(s)
Péptidos y Proteínas de Señalización del Ritmo Circadiano/efectos de los fármacos , Ghrelina/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Páncreas/efectos de los fármacos , ARN Mensajero/efectos de los fármacos , Estómago/efectos de los fármacos , Factores de Transcripción ARNTL/efectos de los fármacos , Factores de Transcripción ARNTL/genética , Animales , Proteínas CLOCK/efectos de los fármacos , Proteínas CLOCK/genética , Caprilatos/farmacología , Péptidos y Proteínas de Señalización del Ritmo Circadiano/genética , Criptocromos/efectos de los fármacos , Criptocromos/genética , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/metabolismo , Regulación de la Expresión Génica , Ghrelina/genética , Ghrelina/metabolismo , Transportador de Glucosa de Tipo 2/efectos de los fármacos , Transportador de Glucosa de Tipo 2/genética , Hipotálamo/metabolismo , Ratones , Nicotina/administración & dosificación , Agonistas Nicotínicos/administración & dosificación , Páncreas/metabolismo , Proteínas Circadianas Period/efectos de los fármacos , Proteínas Circadianas Period/genética
3.
Mol Cell Biol ; 39(21)2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31481450

RESUMEN

Streptozotocin (STZ) is widely used to induce diabetic rodent models. It is specifically toxic to pancreatic beta cells and causes severe destruction and dysfunction. We investigated the effect of 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) on an STZ-induced diabetic mouse model. PLAG attenuated the glucose increase and maintained serum insulin at levels similar to those seen with control mice. In pancreatic beta cell line INS-1, STZ-induced cell apoptosis and intracellular reactive oxygen species (ROS) generation were significantly reduced to nearly normal levels after PLAG treatment. Glucose transporter 2 (GLUT2) localization analyses and glucose uptake assays showed that PLAG accelerated GLUT2 internalization, which ameliorated excessive entry of glucose, as well as STZ. STZ-induced cytotoxic effects were significantly reduced in PLAG-treated groups. The biological activity of PLAG was further confirmed in GLUT2-silenced cells, and the specificity of PLAG was verified using its derivative 1-palmitoyl-2-linoleoyl-3-hydroxyl-rac-glycerol (PLH). Our results suggest that PLAG may be a useful agent for protecting beta cells in the setting of excessive glucose influx.


Asunto(s)
Diglicéridos/farmacología , Transportador de Glucosa de Tipo 2/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Animales , Glucemia/efectos de los fármacos , Línea Celular , Diabetes Mellitus Experimental/tratamiento farmacológico , Diglicéridos/metabolismo , Modelos Animales de Enfermedad , Endocitosis/efectos de los fármacos , Endocitosis/fisiología , Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratas , Estreptozocina
4.
Vet Med Sci ; 5(3): 451-461, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30973212

RESUMEN

The ban on the use of antibiotic in feed encouraged nutritionists to using alternatives to maintain growth performance and intestinal function of broilers. This study was conducted to evaluate the effects of Yupingfeng polysaccharides (YP) supplementation on growth performance and expression of SGLT1, GLUT2 and GLUT5 in Qingyuan partridge chicken. Experiment 1: a total of 540 chickens were randomly allocated to five groups with six replication. Dietary treatments were: (1) CON (control group), basal diet; (2) T1, CON + 0.5 g kg-1 YP; (3) T2, CON + 1 g kg-1 YP; (4) T3, CON + 2 g kg-1 YP; (5) T4, CON + 4 g kg-1 YP. Experiment 2, a total of 162 were randomly allocated to three groups with three replication. Dietary treatments were: (1) CON, basal diet; (2) T1, CON + 0.5 g kg-1 YP; (3) T2, CON + 1 g kg-1 YP. From days 1 to 14 and overall, chicken fed T1 diet had higher ADG. On day 42, there was increased villus height of jejunum in T1 group. On days 14 and 28, there was decreased villus height of duodenum and jejunum in T2 group. In duodenum, the expression of SGLT1 (days 21, 35 and 42), GLUT2 (days 7, 14, 21, 28, 35 and 42) and GLUT5 (days 7, 14, 21 and 28) was increased with YP supplementation. In jejunum, the expression of SGLT1 (days 7, 14, 21, 28 and 35), GLUT2 (days 14, 21, 28, 35 and 42) and GLUT5 (days 7, 14, 21, 28, 35 and 42) was increased with YP supplementation. In ileum, the expression of SGLT1 (days 7, 21, 35 and 42), GLUT2 (days 7, 14, 21 and 42) and GLUT5 (days 7, 14, 21, 28, 35 and 42) was increased with YP supplementation. Dietary YP supplementation improves growth performance and expression of SGLT1, GLUT2 and GLUT5 in intestine.


Asunto(s)
Pollos/crecimiento & desarrollo , Suplementos Dietéticos/análisis , Medicamentos Herbarios Chinos/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Polisacáridos/farmacología , Alimentación Animal/análisis , Animales , Pollos/anatomía & histología , Pollos/genética , Dieta/veterinaria , Regulación del Desarrollo de la Expresión Génica/genética , Transportador de Glucosa de Tipo 2/efectos de los fármacos , Transportador de Glucosa de Tipo 2/genética , Transportador de Glucosa de Tipo 5/efectos de los fármacos , Transportador de Glucosa de Tipo 5/genética , Mucosa Intestinal/anatomía & histología , Mucosa Intestinal/efectos de los fármacos , Intestino Delgado/anatomía & histología , Intestino Delgado/efectos de los fármacos , ARN Mensajero/efectos de los fármacos , ARN Mensajero/genética , Distribución Aleatoria , Transportador 1 de Sodio-Glucosa/efectos de los fármacos , Transportador 1 de Sodio-Glucosa/genética , Regulación hacia Arriba
5.
Stress ; 20(6): 562-572, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28911262

RESUMEN

High ambient temperature adversely influences poultry production. In the present study, gamma amino butyric acid (GABA) supplementation was used to alleviate the adverse changes due to heat stress (HS) in a broiler chicken strain (Ross 308). At 21 days of age, the birds were divided into four groups of 13. Two groups were housed under normal room temperature, one group was given orally 0.2 ml 0.9% physiological saline (CN) daily, the other group received 0.2 ml of 0.5% GABA solution orally (GN). A third group was exposed to environmental HS (33 ± 1 °C lasting for 2 weeks) + physiological saline (CH) and a fourth group was exposed to HS + GABA supplementation (GH). GABA supplementation during HS significantly reduced the birds' increased body temperature (p <.0001) and increased their body weight gain (p <.0001). This effect was associated with increases in the heat stress-induced reductions in jejunal villus length, crypt depth and mucous membrane thickness, and decreases in the vascular changes occurred due to HS. Additionally, GABA supplementation significantly modulated HS-induced changes in glucose facilitated transporter 2 (GLUT2), peptide transporter 1 (PEPT1) and heat shock protein 70 (HSP70) mRNA expression in the jejunal mucosa (p < .0001). GABA supplementation also significantly elevated the triiodothyronine (T3) hormone level and hemoglobin levels and decreased the heterophil-lymphocyte ratio (H/L ratio) (p <.0001). Furthermore, it induced higher hepatic glutathione peroxidase enzyme (GSH-Px) activities and decreased the malondialdehyde dehydrogenase (MDA) content. These results indicate that GABA supplementation during HS may be used to alleviate HS-related changes in broiler chickens.


Asunto(s)
GABAérgicos/farmacología , Transportador de Glucosa de Tipo 2/efectos de los fármacos , Proteínas HSP70 de Choque Térmico/efectos de los fármacos , Respuesta al Choque Térmico/efectos de los fármacos , Calor/efectos adversos , Mucosa Intestinal/efectos de los fármacos , Yeyuno/efectos de los fármacos , Transportador de Péptidos 1/efectos de los fármacos , ARN Mensajero/efectos de los fármacos , Ácido gamma-Aminobutírico/farmacología , Animales , Pollos , Transportador de Glucosa de Tipo 2/genética , Transportador de Glucosa de Tipo 2/metabolismo , Glutatión Peroxidasa/efectos de los fármacos , Glutatión Peroxidasa/metabolismo , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Trastornos de Estrés por Calor , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Yeyuno/metabolismo , Yeyuno/patología , Hígado/efectos de los fármacos , Masculino , Malondialdehído/metabolismo , Oxidorreductasas/efectos de los fármacos , Oxidorreductasas/metabolismo , Transportador de Péptidos 1/genética , Transportador de Péptidos 1/metabolismo , ARN Mensajero/metabolismo , Estrés Fisiológico/efectos de los fármacos
6.
Molecules ; 22(7)2017 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-28708105

RESUMEN

Insulin resistance participates in the glycaemic control disruption in type 2 diabetes mellitus (T2DM), by reducing muscle glucose influx and increasing liver glucose efflux. GLUT4 (Slc2a4 gene) and GLUT2 (Slc2a2 gene) proteins play a fundamental role in the muscle and liver glucose fluxes, respectively. Resveratrol is a polyphenol suggested to have an insulin sensitizer effect; however, this effect, and related mechanisms, have not been clearly demonstrated in T2DM. We hypothesized that resveratrol can improve glycaemic control by restoring GLUT4 and GLUT2 expression in muscle and liver. Mice were rendered obese T2DM in adult life by neonatal injection of monosodium glutamate. Then, T2DM mice were treated with resveratrol for 60 days or not. Glycaemic homeostasis, GLUT4, GLUT2, and SIRT1 (sirtuin 1) proteins (Western blotting); Slc2a4, Slc2a2, and Pck1 (key gluconeogenic enzyme codifier) mRNAs (RT-qPCR); and hepatic glucose efflux were analysed. T2DM mice revealed: high plasma concentration of glucose, fructosamine, and insulin; insulin resistance (insulin tolerance test); decreased Slc2a4/GLUT4 content in gastrocnemius and increased Slc2a2/GLUT2 content in liver; and increased Pck1 mRNA and gluconeogenic activity (pyruvate tolerance test) in liver. All alterations were restored by resveratrol treatment. Additionally, in both muscle and liver, resveratrol increased SIRT1 nuclear content, which must participate in gene expression regulations. In sum, the results indisputably reveals that resveratrol improves glycaemic control in T2DM, and that involves an increase in muscle Slc2a4/GLUT4 and a decrease in liver Slc2a2/GLUT2 expression. This study contributes to our understanding how resveratrol might be prescribed for T2DM according to the principles of evidence-based medicine.


Asunto(s)
Glucemia/efectos de los fármacos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Regulación de la Expresión Génica/efectos de los fármacos , Estilbenos/metabolismo , Estilbenos/farmacología , Animales , Glucosa/metabolismo , Transportador de Glucosa de Tipo 2/efectos de los fármacos , Transportador de Glucosa de Tipo 2/genética , Transportador de Glucosa de Tipo 4/efectos de los fármacos , Transportador de Glucosa de Tipo 4/genética , Humanos , Insulina/metabolismo , Resistencia a la Insulina , Hígado/metabolismo , Masculino , Ratones , Ratones Obesos , Músculo Esquelético/metabolismo , ARN Mensajero/efectos de los fármacos , Resveratrol , Sirtuina 1
7.
Biomed Pharmacother ; 84: 230-236, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27662473

RESUMEN

P-coumaric acid (p-CA, 3-[4-hydroxyphenyl]-2-propenoic acid), the major component widely found in nutritious plant foods, has various antioxidant, antiinflammatory and anticancer property. To evaluate the antidiabetic and antihyperlipidemic mechanisms, via the effects on carbohydrate, lipids and lipoproteins responses in adult male albino Wistar rats were examined by treated with p-CA. Rats were injected with streptozotocin (STZ, 40mg/kg b.w.) by intraperitonially (i.p.) 30days for the induction of experimental diabetes mellitus. Diabetic rats were treated with p-CA orally at a dose of 100mg/kg b.w. The potential defending character of p-CA against diabetic rats was evaluated by performing the various biochemical parameters and glucose transporter such as GLUT2 mRNA expression of pancreas. Administration of p-CA significantly lowers the blood glucose level, gluconeogenic enzymes such as glucose-6-phosphatase and fructose-1,6-bisphosphatase whereas increases the activities of hexokinase, glucose-6 phosphatase dehydrogenase and GSH via by increasing level of insulin. p-CA reduces the total cholesterol and triglycerides in both plasma and tissues i.e. liver and kidney. p-CA also decreases the LDL-C, VLDL-C and it considerably increase the level of HDL-C. A significant decreased expression of GLUT 2 mRNA in the pancreas was recorded in the supplementation of p-CA treated groups. Taken together, these results suggest that p-CA modulates glucose and lipid metabolism via GLUT 2 activation in the pancreatic and has potentially beneficial effects in improving or treating metabolic disorders.


Asunto(s)
Glucemia/efectos de los fármacos , Ácidos Cumáricos/farmacología , Diabetes Mellitus Experimental/tratamiento farmacológico , Transportador de Glucosa de Tipo 2/efectos de los fármacos , Hipoglucemiantes/farmacología , Hipolipemiantes/farmacología , Lípidos/sangre , Páncreas/efectos de los fármacos , Animales , Biomarcadores/sangre , Glucemia/metabolismo , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/inducido químicamente , Regulación hacia Abajo , Transportador de Glucosa de Tipo 2/genética , Transportador de Glucosa de Tipo 2/metabolismo , Glutatión/metabolismo , Insulina/sangre , Riñón/efectos de los fármacos , Riñón/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Páncreas/metabolismo , Propionatos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Wistar , Estreptozocina
8.
World J Gastroenterol ; 22(25): 5769-79, 2016 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-27433090

RESUMEN

AIM: To investigated the effects of urotensin II (UII) on hepatic insulin resistance in HepG2 cells and the potential mechanisms involved. METHODS: Human hepatoma HepG2 cells were cultured with or without exogenous UII for 24 h, in the presence or absence of 100 nmol/L insulin for the last 30 min. Glucose levels were detected by the glucose-oxidase method and glycogen synthesis was analyzed by glycogen colorimetric/fluorometric assay. Reactive oxygen species (ROS) levels were detected with a multimode reader using a 2',7'-dichlorofluorescein diacetate probe. The protein expression and phosphorylation levels of c-Jun N-terminal kinase (JNK), insulin signal essential molecules such as insulin receptor substrate -1 (IRS-1), protein kinase B (Akt), glycogen synthase kinase-3ß (GSK-3ß), and glucose transporter-2 (Glut 2), and NADPH oxidase subunits such as gp91(phox), p67(phox), p47(phox), p40(phox), and p22(phox) were evaluated by Western blot. RESULTS: Exposure to 100 nmol/L UII reduced the insulin-induced glucose consumption (P < 0.05) and glycogen content (P < 0.01) in HepG2 cells compared with cells without UII. UII also abolished insulin-stimulated protein expression (P < 0.01) and phosphorylation of IRS-1 (P < 0.05), associated with down-regulation of Akt (P < 0.05) and GSK-3ß (P < 0.05) phosphorylation levels, and the expression of Glut 2 (P < 0.001), indicating an insulin-resistance state in HepG2 cells. Furthermore, UII enhanced the phosphorylation of JNK (P < 0.05), while the activity of JNK, insulin signaling, such as total protein of IRS-1 (P < 0.001), phosphorylation of IRS-1 (P < 0.001) and GSK-3ß (P < 0.05), and glycogen synthesis (P < 0.001) could be reversed by pretreatment with the JNK inhibitor SP600125. Besides, UII markedly improved ROS generation (P < 0.05) and NADPH oxidase subunit expression (P < 0.05). However, the antioxidant/NADPH oxidase inhibitor apocynin could decrease UII-induced ROS production (P < 0.05), JNK phosphorylation (P < 0.05), and insulin resistance (P < 0.05) in HepG2 cells. CONCLUSION: UII induces insulin resistance, and this can be reversed by JNK inhibitor SP600125 and antioxidant/NADPH oxidase inhibitor apocynin targeting the insulin signaling pathway in HepG2 cells.


Asunto(s)
Resistencia a la Insulina , NADPH Oxidasas/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Urotensinas/farmacología , Acetofenonas/farmacología , Western Blotting , Regulación hacia Abajo , Inhibidores Enzimáticos/farmacología , Glucosa/metabolismo , Transportador de Glucosa de Tipo 2/efectos de los fármacos , Transportador de Glucosa de Tipo 2/metabolismo , Glucógeno/biosíntesis , Glucógeno Sintasa Quinasa 3 beta/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células Hep G2 , Humanos , Hipoglucemiantes/farmacología , Insulina/farmacología , Proteínas Sustrato del Receptor de Insulina/efectos de los fármacos , Proteínas Sustrato del Receptor de Insulina/metabolismo , MAP Quinasa Quinasa 4/efectos de los fármacos , MAP Quinasa Quinasa 4/metabolismo , NADPH Oxidasas/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo
9.
J Agric Food Chem ; 63(51): 10903-13, 2015 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-26592089

RESUMEN

Cichoric acid, a caffeic acid derivative found in Echinacea purpurea, basil, and chicory, has been reported to have bioactive effects, such as anti-inflammatory, antioxidant, and preventing insulin resistance. In this study, to explore the effects of CA on regulating insulin resistance and chronic inflammatory responses, the insulin resistance model was constructed by glucosamine in HepG2 cells. CA stimulated glucosamine-mediated glucose uptake by stimulating translocation of the glucose transporter 2. Moreover, the production of reactive oxygen, the expression of COX-2 and iNOS, and the mRNA levels of TNF-α and IL-6 were attenuated. Furthermore, CA was verified to promote glucosamine-mediated glucose uptake and inhibited inflammation through PI3K/Akt, NF-κB, and MAPK signaling pathways in HepG2 cells. These results implied that CA could increase glucose uptake, improve insulin resistance, and attenuate glucosamine-induced inflammation, suggesting that CA is a potential natural nutraceutical with antidiabetic properties and anti-inflammatory effects.


Asunto(s)
Antiinflamatorios , Ácidos Cafeicos/farmacología , Glucosamina/farmacología , Hipoglucemiantes , Inflamación/prevención & control , Resistencia a la Insulina , Succinatos/farmacología , Suplementos Dietéticos , Glucosa/metabolismo , Transportador de Glucosa de Tipo 2/efectos de los fármacos , Transportador de Glucosa de Tipo 2/metabolismo , Células Hep G2 , Humanos , Inflamación/inducido químicamente , Hígado/efectos de los fármacos , Hígado/metabolismo , Transducción de Señal/efectos de los fármacos
10.
Eur J Pharmacol ; 761: 391-7, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25956617

RESUMEN

GLUT2 is a bidirectional glucose transporter present in liver, kidney and pancreas. Studies have shown over-expression of GLUT2 in diabetic conditions. Ferulic acid (FA) is an antidiabetic phenolic phytocompound which is reported to regulate GLUT4 in vitro. The objective of our study is to evaluate the role of FA in the regulation of hepatic GLUT2 expression and the underlying mechanism. Male Wistar rats were divided into 5 groups: control, diabetic (diabetes was induced by giving high fat diet and high fructose water for 60 days), diabetic rats treated with FA (50mg/kg body weight/day, orally for 30 days), diabetic rats treated with metformin (50mg/kg body weight/day, orally for 30 days) and control rats treated with FA (50mg/kg body weight/day orally for 30 days). After 30 days treatment, animals were perfused and liver was dissected out. Glucose uptake and oxidation, expression of GLUT2 and binding of transcription factors - SREBP1c, HNF1α and HNF3ß with GLUT2 gene promoter were studied. Over-expression of GLUT2 in hepatic tissue was found in high fat and fructose- induced type-2 diabetic animals. FA treatment reduced the GLUT2 expression in diabetic animals by impairing the interaction between these transcription factors (SREBP1c, HNF1α and HNF3ß) and GLUT2 gene promoter.


Asunto(s)
Ácidos Cumáricos/farmacología , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Dieta Alta en Grasa , Fructosa , Transportador de Glucosa de Tipo 2/efectos de los fármacos , Hipoglucemiantes/farmacología , Hígado/efectos de los fármacos , Animales , Sitios de Unión , Desoxiglucosa/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Transportador de Glucosa de Tipo 2/genética , Transportador de Glucosa de Tipo 2/metabolismo , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Factor Nuclear 3-beta del Hepatocito/metabolismo , Hígado/metabolismo , Masculino , Oxidación-Reducción , Regiones Promotoras Genéticas , Ratas Wistar , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo
11.
Int J Biol Sci ; 11(5): 508-24, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25892959

RESUMEN

Diabetes mellitus (DM) is a metabolic diseases characterized by hyperglycemia due to insufficient or inefficient insulin secretory response. This chronic disease is a global problem and there is a need for greater emphasis on therapeutic strategies in the health system. Phytochemicals such as flavonoids have recently attracted attention as source materials for the development of new antidiabetic drugs or alternative therapy for the management of diabetes and its related complications. The antidiabetic potential of flavonoids are mainly through their modulatory effects on glucose transporter by enhancing GLUT-2 expression in pancreatic ß cells and increasing expression and promoting translocation of GLUT-4 via PI3K/AKT, CAP/Cb1/TC10 and AMPK pathways. This review highlights the recent findings on beneficial effects of flavonoids in the management of diabetes with particular emphasis on the investigations that explore the role of these compounds in modulating glucose transporter proteins at cellular and molecular level.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Dieta/veterinaria , Flavonoides/farmacología , Transportador de Glucosa de Tipo 2/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Suplementos Dietéticos , Flavonoides/química , Transportador de Glucosa de Tipo 2/efectos de los fármacos , Transportador de Glucosa de Tipo 4/efectos de los fármacos , Humanos , Hipoglucemiantes/química , Hipoglucemiantes/farmacología , Fitoquímicos
12.
Eur Rev Med Pharmacol Sci ; 18(9): 1287-94, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24867506

RESUMEN

AIM: Type 2 diabetes mellitus is characterized by lack of, or relative deficiency in, insulin productions and insensitivity of target tissues to insulin. Improvement of ß-cell functions is a potential strategy for the clinical management of this disease. We reported before that geniposide improved glucose-stimulated insulin secretion with the activation of glucagon-like peptide 1 receptor (GLP-1R) in INS-1 pancreatic ß cells, but the cell signaling mechanism of geniposide regulating glucose-stimulated insulin secretion (GSIS) in ß cells is so far poorly understood. MATERIALS AND METHODS: Effect of LY294002, a specific inhibitor of PI3K, on GSIS in the presence or absence of geniposide in INS-1 cells. In addition, the differential protein expression of geniposide treated INS-1 cells was examined by Western blot. RESULTS: After pretreatment with 10 µM LY294002 for 1 hour, the insulin secretion induced by geniposide was partly abolished in INS-1 cells. After treatment with geniposide, the phosphorylation of PDK1 and Akt473 increased gradually to the maximum at 60 minutes or 120 minutes respectively. Furthermore, geniposide also inhibited the phosphorylation of downstream target GSK3ß, and this effect was counteracted by preincubation with LY294002. And the expression of GLUT2 was increased after treatment with different doses geniposide. CONCLUSIONS: Geniposide increases insulin secretion in pancreatic ß cells in a PI3K dependent mechanism potentially through increased GLUT2 protein levels.


Asunto(s)
Hipoglucemiantes/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Insulina/metabolismo , Iridoides/farmacología , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Animales , Línea Celular , Receptor del Péptido 1 Similar al Glucagón , Glucosa/metabolismo , Transportador de Glucosa de Tipo 2/efectos de los fármacos , Transportador de Glucosa de Tipo 2/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Secreción de Insulina , Células Secretoras de Insulina/enzimología , Células Secretoras de Insulina/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Receptores de Glucagón/agonistas , Receptores de Glucagón/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Regulación hacia Arriba
13.
Am J Physiol Regul Integr Comp Physiol ; 305(7): R840-53, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23926132

RESUMEN

Post-oral sugar actions enhance the intake of and preference for sugar-rich foods, a process referred to as appetition. Here, we investigated the role of intestinal sodium glucose cotransporters (SGLTs) in sugar appetition in C57BL/6J mice using sugars and nonmetabolizable sugar analogs that differ in their affinity for SGLT1 and SGLT3. In experiments 1 and 2, food-restricted mice were trained (1 h/day) to consume a flavored saccharin solution [conditioned stimulus (CS-)] paired with intragastric (IG) self-infusions of water and a different flavored solution (CS+) paired with infusions of 8 or 12% sugars (glucose, fructose, and galactose) or sugar analogs (α-methyl-D-glucopyranoside, MDG; 3-O-methyl-D-glucopyranoside, OMG). Subsequent two-bottle CS+ vs. CS- choice tests were conducted without coinfusions. Infusions of the SGLT1 ligands glucose, galactose, MDG, and OMG stimulated CS+ licking above CS- levels. However, only glucose, MDG, and galactose conditioned significant CS+ preferences, with the SGLT3 ligands (glucose, MDG) producing the strongest preferences. Fructose, which is not a ligand for SGLTs, failed to stimulate CS+ intake or preference. Experiment 3 revealed that IG infusion of MDG+phloridzin (an SGLT1/3 antagonist) blocked MDG appetition, whereas phloridzin had minimal effects on glucose-induced appetition. However, adding phloretin (a GLUT2 antagonist) to the glucose+phloridzin infusion blocked glucose appetition. Taken together, these findings suggest that humoral signals generated by intestinal SGLT1 and SGLT3, and to a lesser degree, GLUT2, mediate post-oral sugar appetition in mice. The MDG results indicate that sugar metabolism is not essential for the post-oral intake-stimulating and preference-conditioning actions of sugars in mice.


Asunto(s)
Regulación del Apetito/efectos de los fármacos , Carbohidratos/administración & dosificación , Preferencias Alimentarias/efectos de los fármacos , Administración Oral , Animales , Condicionamiento Psicológico/efectos de los fármacos , Ingestión de Líquidos/efectos de los fármacos , Fructosa/administración & dosificación , Fructosa/metabolismo , Galactosa/administración & dosificación , Galactosa/metabolismo , Glucosa/administración & dosificación , Glucosa/metabolismo , Transportador de Glucosa de Tipo 2/efectos de los fármacos , Transportador de Glucosa de Tipo 2/metabolismo , Absorción Intestinal , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Ligandos , Masculino , Metilglucósidos/administración & dosificación , Metilglucósidos/metabolismo , Ratones , Ratones Endogámicos C57BL , Sacarina/administración & dosificación , Autoadministración , Proteínas de Transporte de Sodio-Glucosa/efectos de los fármacos , Proteínas de Transporte de Sodio-Glucosa/metabolismo , Transportador 1 de Sodio-Glucosa/efectos de los fármacos , Transportador 1 de Sodio-Glucosa/metabolismo , Edulcorantes/administración & dosificación , Factores de Tiempo
14.
Diabetologia ; 50(10): 2209-17, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17694297

RESUMEN

AIMS/HYPOTHESIS: GLUT2 is the main renal glucose transporter upregulated by hyperglycaemia, when it becomes detectable at the brush border membrane (BBM). Since glucose-induced protein kinase C (PKC) activation in the kidney is linked to diabetic nephropathy, we investigated the effect of glycaemic status on the protein levels of PKC isoforms alpha, betaI, betaII, delta and epsilon in the proximal tubule, as well as the relationship between them and changes in GLUT2 production at the BBM. METHODS: Plasma glucose concentrations were modulated in rats by treatment with nicotinamide 15 min prior to induction of diabetes with streptozotocin. Levels of GLUT2 protein and PKC isoforms in BBM were measured by western blotting. Additionally, the role of calcium signalling and PKC activation on facilitative glucose transport was examined by measuring glucose uptake in BBM vesicles prepared from proximal tubules that had been incubated either with thapsigargin, which increases cytosolic calcium, or with the PKC activator phorbol 12-myristate,13-acetate (PMA). RESULTS: Thapsigargin and PMA enhanced GLUT-mediated glucose uptake, but had no effect on sodium-dependent glucose transport. Diabetes significantly increased the protein levels of GLUT2 and PKC-betaI at the BBM. Levels of GLUT2 and PKC-betaI correlated positively with plasma glucose concentration. Diabetes had no effect on BBM levels of alpha, betaII, delta or epsilon isoforms of PKC. CONCLUSIONS/INTERPRETATION: Enhanced GLUT2-mediated glucose transport across the proximal tubule BBM during diabetic hyperglycaemia is closely associated with increased PKC-betaI. Thus, altered levels of GLUT2 and PKC-betaI proteins in the BBM may be important factors in the pathogenic processes underlying diabetic renal injury.


Asunto(s)
Glucemia/metabolismo , Diabetes Mellitus Experimental/metabolismo , Transportador de Glucosa de Tipo 2/metabolismo , Túbulos Renales Proximales/metabolismo , Microvellosidades/metabolismo , Proteína Quinasa C/metabolismo , Animales , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/enzimología , Activación Enzimática , Transportador de Glucosa de Tipo 2/efectos de los fármacos , Riñón/anatomía & histología , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/fisiología , Masculino , Microvellosidades/efectos de los fármacos , Microvellosidades/fisiología , Niacinamida/farmacología , Tamaño de los Órganos , Proteína Quinasa C/efectos de los fármacos , Proteína Quinasa C beta , Ratas , Ratas Sprague-Dawley , Acetato de Tetradecanoilforbol/farmacología , Tapsigargina/farmacología , Aumento de Peso
15.
Life Sci ; 79(2): 144-53, 2006 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-16426640

RESUMEN

The effects of aporphines and secoaporphines on glucose uptake by isolated intestinal brush-border membrane vesicles (BBMV) or basolateral membrane vesicles (BLMV) and glucose absorption during in situ intestinal perfusion were studied. Of the tested compounds, N-allylsecoboldine was the most potent glucose uptake inhibitor, with IC50 values of 159 microM and 121 microM, respectively, for uptake by BBMV and BLMV. While thaliporphine competitively inhibited glucose uptake by both membrane preparations, inhibition by N-allylsecoboldine was competitive using BBMV and noncompetitive using BLMV. In addition, N-allylsecoboldine significantly reduced both glucose absorption during in situ intestinal perfusion and blood glucose levels in the oral glucose tolerance test. The results demonstrate that levels of both aporphines and secoaporphines achievable by oral administration have an inhibitory effect on intestinal glucose uptake and suggest that the hypoglycemic effects of these compounds merit attention.


Asunto(s)
Aporfinas/farmacología , Glucosa/metabolismo , Mucosa Intestinal/metabolismo , Algoritmos , Animales , Relación Dosis-Respuesta a Droga , Prueba de Tolerancia a la Glucosa , Transportador de Glucosa de Tipo 2/efectos de los fármacos , Transportador de Glucosa de Tipo 2/metabolismo , Absorción Intestinal/efectos de los fármacos , Masculino , Microvellosidades/metabolismo , Ratas , Ratas Wistar , Vesículas Secretoras/metabolismo , Transportador 1 de Sodio-Glucosa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...