Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Int J Mol Sci ; 25(5)2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38473959

ABSTRACT

Advanced glycation end products (AGEs) prime macrophages for lipopolysaccharide (LPS)-induced inflammation. We investigated the persistence of cellular AGE-sensitization to LPS, considering the nuclear content of p50 and p65 nuclear factor kappa B (NFKB) subunits and the expression of inflammatory genes. Macrophages treated with control (C) or AGE-albumin were rested for varying intervals in medium alone before being incubated with LPS. Comparisons were made using one-way ANOVA or Student t-test (n = 6). AGE-albumin primed macrophages for increased responsiveness to LPS, resulting in elevated levels of TNF, IL-6, and IL-1beta (1.5%, 9.4%, and 5.6%, respectively), compared to C-albumin. TNF, IL-6, and IL-1 beta secretion persisted for up to 24 h even after the removal of AGE-albumin (area under the curve greater by 1.6, 16, and 5.2 times, respectively). The expressions of Il6 and RelA were higher 8 h after albumin removal, and Il6 and Abca1 were higher 24 h after albumin removal. The nuclear content of p50 remained similar, but p65 showed a sustained increase (2.9 times) for up to 24 h in AGE-albumin-treated cells. The prolonged activation of the p65 subunit of NFKB contributes to the persistent effect of AGEs on macrophage inflammatory priming, which could be targeted for therapies to prevent complications based on the AGE-RAGE-NFKB axis.


Subject(s)
Interleukin-6 , NF-kappa B , NF-kappa B/metabolism , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Glycation End Products, Advanced/metabolism , Albumins/metabolism
2.
Life Sci ; 287: 120143, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34785192

ABSTRACT

AIMS: To investigate the effect of resistance training-RT on glycemia, expression of the glucose transporter-GLUT4, bone mineral density-BMD, and microstructural and biomechanical properties of osteopenic rat bones in neonatal streptozotocin-induced diabetes. MAIN METHODS: Sixty-four 5-day-old male rats were divided into two groups: control and diabetic rats injected with vehicle or streptozotocin, respectively. After 55 days, densitometric analysis-DA of the tibia was performed. These groups were subdivided into four subgroups: non-osteopenic control-CN, osteopenic control-OC, non-osteopenic diabetic-DM, and osteopenic diabetic-OD. The OC and OD groups were suspended by their tails for 21 days to promote osteopenia in the hindlimb; subsequently, a second DA was performed. The rats were subdivided into eight subgroups: sedentary control-SC, sedentary osteopenic control-SOC, exercised control-EC, exercised osteopenic control-EOC, sedentary diabetic-SD, sedentary osteopenic diabetic-SOD, exercised diabetic-ED, and exercised osteopenic diabetic-EOD. For RT, the rats climbed a ladder with weights secured to their tails for 12 weeks. After RT, a third DA was performed, and blood samples, muscles, and tibias were assessed to measure glycemia, insulinemia, GLUT4 content, bone maximum strength, fracture energy, extrinsic stiffness, BMD, cancellous bone area, trabecular number, and trabecular width. KEY FINDINGS: After RT, glycemia, GLUT4 content, BMD, and bone microstructural and biomechanical properties were improved in diabetic rats (osteopenic and non-osteopenic). However, RT had no effect on these parameters in the EC and SC groups. SIGNIFICANCE: These results suggest that RT improves GLUT4 content, BMD, and microstructural and biomechanical properties of bone in osteopenic and non-osteopenic diabetic rats and is effective in controlling glycemia.


Subject(s)
Biomechanical Phenomena/physiology , Bone Density/physiology , Bone Diseases, Metabolic/metabolism , Diabetes Mellitus, Experimental/metabolism , Glucose Transporter Type 4/metabolism , Resistance Training/methods , Animals , Bone Diseases, Metabolic/diagnostic imaging , Bone Diseases, Metabolic/therapy , Diabetes Mellitus, Experimental/diagnostic imaging , Diabetes Mellitus, Experimental/therapy , Male , Physical Conditioning, Animal/methods , Physical Conditioning, Animal/physiology , Rats , Rats, Wistar
3.
J Biol Chem ; 296: 100344, 2021.
Article in English | MEDLINE | ID: mdl-33524391

ABSTRACT

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.


Subject(s)
Diet, Sodium-Restricted , Insulin Resistance , Lipid Metabolism , Muscle, Skeletal/metabolism , Animals , Lipidomics , Male , Mice , Sodium, Dietary/metabolism
4.
Mol Cell Endocrinol ; 481: 26-34, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30528377

ABSTRACT

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.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/metabolism , Glucose Transporter Type 4/genetics , Histones/metabolism , Muscle, Skeletal/metabolism , Animals , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 2/genetics , Disease Models, Animal , Epigenesis, Genetic , Glucose Transporter Type 4/metabolism , Histones/chemistry , Humans , Insulin , Lysine/metabolism , Male , Methylation , Mice , Promoter Regions, Genetic , Rats , Resveratrol
5.
Sci Rep ; 8(1): 8109, 2018 05 25.
Article in English | MEDLINE | ID: mdl-29802324

ABSTRACT

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.


Subject(s)
Gene Expression Regulation/drug effects , Glucose Transporter Type 4/genetics , Glycation End Products, Advanced/pharmacology , Insulin Resistance , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Animals , Biomarkers/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Glucose/metabolism , Homeostasis/drug effects , Male , NF-kappa B/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar
6.
Front Physiol ; 8: 723, 2017.
Article in English | MEDLINE | ID: mdl-29018354

ABSTRACT

Background: Advanced glycation endproducts elicit inflammation. However, their role in adipocyte macrophage infiltration and in the development of insulin resistance, especially in the absence of the deleterious biochemical pathways that coexist in diabetes mellitus, remains unknown. We investigated the effect of chronic administration of advanced glycated albumin (AGE-albumin) in healthy rats, associated or not with N-acetylcysteine (NAC) treatment, on insulin sensitivity, adipose tissue transcriptome and macrophage infiltration and polarization. Methods: Male Wistar rats were intraperitoneally injected with control (C) or AGE-albumin alone, or, together with NAC in the drinking water. Biochemical parameters, lipid peroxidation, gene expression and protein contents were, respectively, determined by enzymatic techniques, reactive thiobarbituric acid substances, RT-qPCR and immunohistochemistry or immunoblot. Carboxymethyllysine (CML) and pyrraline (PYR) were determined by LC/mass spectrometry (LC-MS/MS) and ELISA. Results: CML and PYR were higher in AGE-albumin as compared to C. Food consumption, body weight, systolic blood pressure, plasma lipids, glucose, hepatic and renal function, adipose tissue relative weight and adipocyte number were similar among groups. In AGE-treated animals, insulin resistance, adipose macrophage infiltration and Col12a1 mRNA were increased with no changes in M1 and M2 phenotypes as compared to C-albumin-treated rats. Total GLUT4 content was reduced by AGE-albumin as compared to C-albumin. NAC improved insulin sensitivity, reduced urine TBARS, adipose macrophage number and Itgam and Mrc mRNA and increased Slc2a4 and Ppara. CD11b, CD206, Ager, Ddost, Cd36, Nfkb1, Il6, Tnf, Adipoq, Retn, Arg, and Il12 expressions were similar among groups. Conclusions: AGE-albumin sensitizes adipose tissue to inflammation due to macrophage infiltration and reduces GLUT4, contributing to insulin resistance in healthy rats. NAC antagonizes AGE-albumin and prevents insulin resistance. Therefore, it may be a useful tool in the prevention of AGE action on insulin resistance and long-term complications of DM.

7.
Mol Cell Endocrinol ; 447: 116-124, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28238722

ABSTRACT

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.


Subject(s)
Glycation End Products, Advanced/pharmacology , Hormesis/drug effects , Insulin Resistance , Liver/metabolism , Albumins/pharmacology , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Gene Expression Regulation/drug effects , Gluconeogenesis/drug effects , Gluconeogenesis/genetics , Glycation End Products, Advanced/administration & dosage , Glycogen/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , HMGB1 Protein/metabolism , Inflammation Mediators/metabolism , Injections, Intraperitoneal , Lipogenesis/drug effects , Lipogenesis/genetics , Liver/drug effects , Male , Models, Biological , NF-E2-Related Factor 2/metabolism , Oxidation-Reduction , Proto-Oncogene Proteins c-akt/metabolism , Rats, Wistar , Reactive Oxygen Species/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism
8.
Nutr Metab (Lond) ; 13: 44, 2016.
Article in English | MEDLINE | ID: mdl-27366200

ABSTRACT

BACKGROUND: Resveratrol is a natural polyphenol that has been proposed to improve glycemic control in diabetes, by mechanisms that involve improvement in insulin secretion and activity. In type 1 diabetes (T1D), in which insulin therapy is obligatory, resveratrol treatment has never been investigated. The present study aimed to evaluate resveratrol as an adjunctive agent to insulin therapy in a T1D-like experimental model. METHODS: Rats were rendered diabetic by streptozotocin (STZ) treatment. Twenty days later, four groups of animals were studied: non-diabetic (ND); diabetic treated with placebo (DP); diabetic treated with insulin (DI) and diabetic treated with insulin plus resveratrol (DIR). After 30 days of treatment, 24-hour urine was collected; then, blood, soleus muscle, proximal small intestine, renal cortex and liver were sampled. Specific glucose transporter proteins were analyzed (Western blotting) in each territory of interest. Solute carrier family 2 member 2 (Slc2a2), phosphoenolpyruvate carboxykinase (Pck1) and glucose-6-phosphatase catalytic subunit (G6pc) mRNAs (qPCR), glycogen storage and sirtuin 1 (SIRT1) activity were analyzed in liver. RESULTS: Diabetes induction increased blood glucose, plasma fructosamine concentrations, and glycosuria. Insulin therapy partially recovered the glycemic control; however, resveratrol as adjunctive therapy additionally improved glycemic control and restored plasma fructosamine concentration to values of non-diabetic rats. Resveratrol did not alter the expression of the glucose transporters GLUT2 and SGLT1 in the intestine, GLUT2 and SGLT2 in kidney and GLUT4 in soleus, suggesting that fluxes of glucose in these territories were unaltered. Differently, in liver, resveratrol promoted a reduction in Slc2a2, Pck1, and G6pc mRNAs, as well as in GLUT2 protein (P < 0.05, DIR vs. DI); besides, it increased (P < 0.01, DIR vs. DI) the hepatic glycogen content, and SIRT1 protein. CONCLUSIONS: Resveratrol is able to improve glycemic control in insulin-treated T1D-like rats. This effect seems not to involve changes in glucose fluxes in the small intestine, renal proximal tubule, and soleus skeletal muscle; but to be related to several changes in the liver, where downregulation of Slc2a2/GLUT2, Pck1, and G6pc expression was observed, favoring reduction of glucose production and efflux. Besides, resveratrol increased SIRT1 nuclear protein content in liver, which may be related to the observed gene expression regulations.

9.
Life Sci ; 148: 194-200, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26854998

ABSTRACT

AIMS: The fetal programming hypothesis suggests that intrauterine stimuli can induce metabolic changes in offspring, increasing the disease risk in adulthood. Periodontal disease may enhance serum cytokine levels. Cytokines such as tumor necrosis factor-alpha (TNF-α) have been associated with reduced glucose transporter type 4 (GLUT4) expression, decreased protein kinase B (Akt) phosphorylation, and insulin resistance. This study aimed to evaluate GLUT4 content, and Akt serine phosphorylation status in the gastrocnemius skeletal muscle (GSM), glycemia, insulinemia and change in body weight in offspring of rats with periodontal disease. MAIN METHODS: Female Wistar rats were distributed into a control group (CN) and an experimental periodontal disease group (PD), in which a ligature was placed around the mandibular first molars. Seven days after ligature placement, both groups were mated with normal male rats. The ligatures remained throughout pregnancy until weaning, after which the male offspring were distributed into groups: CN-o, control rat offspring; and PD-o, periodontal disease rat offspring. The body weight from 0 to 75days of age was measured. At 75days, the glycemia, insulinemia, TNF-α levels, Akt serine phosphorylation, and GLUT4 content in the GSM were measured in the offspring. KEY FINDINGS: The PD-o group showed a low birth weight (LBW), unchanged glycemia, increased insulinemia, insulin resistance, increased TNF-α levels, decreased Akt serine phosphorylation status, and reduced GLUT4 content in the plasma membrane and translocation index after insulin stimulation. SIGNIFICANCE: Maternal periodontal disease causes LBW, insulin resistance, and alterations in the final stage of insulin signaling in the GSM of adult offspring.


Subject(s)
Cell Membrane/metabolism , Glucose Transporter Type 4/metabolism , Muscle, Skeletal/metabolism , Pregnancy Complications/metabolism , Animals , Blood Glucose/metabolism , Female , Insulin Resistance/physiology , Male , Pregnancy , Pregnancy Complications/pathology , Rats , Rats, Wistar
10.
Clin Oral Investig ; 20(7): 1625-30, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26592809

ABSTRACT

OBJECTIVES: Periapical lesion (PL) promotes insulin resistance; however, the mechanisms underlying this alteration are not fully understood. Therefore, in this study, we aimed to evaluate the Akt serine phosphorylation status and GLUT4 expression levels in the gastrocnemius muscle (GM) of rats with PL. MATERIALS AND METHODS: Male Wistar rats (n = 42) were distributed equally into control (CN) and PL groups. The pulpal tissue of the PL group rats was exposed to the oral environment for 30 days. Thereafter, glucose and insulin levels were assessed, followed by homeostasis model assessment of insulin resistance (HOMA-IR). The Akt serine phosphorylation and GLUT4 levels of microsomal (M) and plasma membrane (PM) fractions were evaluated by western blotting and analyzed statistically. RESULTS: Compared to CN group rats, PL group rats had lower insulin sensitivity (as observed by HOMA-IR), lower Akt serine phosphorylation status after insulin stimulus, and lower GLUT4 levels in the PM fraction. However, the M fraction in the PL group did not differ significantly from that of the CN group. CONCLUSIONS: PL decreases insulin sensitivity, Akt phosphorylation, and PM GLUT4 content. CLINICAL RELEVANCE: The present study indicates that preventing endodontic disease can thwart insulin resistance.


Subject(s)
Dental Pulp/injuries , Glucose Transporter Type 4/metabolism , Muscle, Skeletal/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Blotting, Western , Cell Membrane/metabolism , Disease Models, Animal , Insulin Resistance , Male , Phosphorylation , Rats , Rats, Wistar
11.
Biomed Res Int ; 2015: 282984, 2015.
Article in English | MEDLINE | ID: mdl-25722973

ABSTRACT

We characterized the metabolic profile of transgenic mice exhibiting enhanced muscle mass driven by increased mIGF-1 expression (MLC/mIGF-1). As expected, 6-month-old MLC/mIGF-1 mice were heavier than age-matched wild type (WT) mice (37.4 ± 0.3 versus 31.8 ± 0.6 g, resp.). MLC/mIGF-1 mice had higher respiratory quotient when compared to WT (0.9 ± 0.03 versus 0.74 ± 0.02, resp.) suggesting a preference for carbohydrate as the major fuel source. MLC/mIGF-1 mice had a higher rate of glucose disposal when compared to WT (3.25 ± 0.14 versus 2.39 ± 0.03%/min, resp.). The higher disposal rate correlated to ∼ 2-fold higher GLUT4 content in the extensor digitorum longus (EDL) muscle. Analysis of mRNA content for the glycolysis-related gene PFK-1 showed ∼ 3-fold upregulation in MLC/mIGF-1 animals. We also found a 50% downregulation of PGC1α mRNA levels in MLC/mIGF-1 mouse EDL muscle, suggesting less abundant mitochondria in this tissue. We found no difference in the expression of PPARα and PPARß/δ, suggesting no modulation of key elements in oxidative metabolism. These data together suggest a shift in metabolism towards higher carbohydrate utilization, and that could explain the increased insulin sensitivity of hypertrophied skeletal muscle in MLC/mIGF-1 mice.


Subject(s)
Carbohydrate Metabolism/physiology , Hypertrophy/metabolism , Insulin Resistance/physiology , Insulin-Like Growth Factor I/metabolism , Insulin/metabolism , Muscle, Skeletal/metabolism , Animals , Glucose Transporter Type 4/metabolism , Mice , Mice, Transgenic , Mitochondria/metabolism , Muscle Proteins/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Peroxisome Proliferator-Activated Receptors/metabolism , RNA, Messenger/metabolism , Transcription Factors/metabolism
12.
Life Sci ; 93(22): 805-13, 2013 Nov 19.
Article in English | MEDLINE | ID: mdl-24157454

ABSTRACT

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.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Glucose Transporter Type 2/genetics , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 4/genetics , Kidney/metabolism , Liver/metabolism , Animals , Binding Sites , CCAAT-Enhancer-Binding Protein-beta/genetics , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Gene Expression Regulation/drug effects , Glucose Transporter Type 2/metabolism , Hepatocyte Nuclear Factor 1-alpha/metabolism , Hepatocyte Nuclear Factor 3-beta/metabolism , Hepatocyte Nuclear Factor 4/metabolism , Insulin/pharmacology , Kidney/drug effects , Liver/drug effects , Male , Promoter Regions, Genetic , Rats , Rats, Wistar , Sterol Regulatory Element Binding Protein 1/genetics
13.
Diabetol Metab Syndr ; 5(1): 64, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24499577

ABSTRACT

BACKGROUND: Oral health complications in diabetes and hypertension include decreased salivary secretion. The sodium-glucose cotransporter 1 (SGLT1) protein, which transports 1 glucose/2 Na+/264 H2O molecules, is described in salivary glands. We hypothesized that changes in SGLT1 expression in the luminal membrane of ductal cell may be related to an altered salivary flow. FINDINGS: By immunohistochemistry, we investigated SGLT1 expression in ductal cells of parotid and submandibular glands from Wistar Kyoto rats (WKY), diabetic WKY (WKY-D), spontaneously hypertensive rats (SHR) and diabetic SHR (SHR-D), as well as in parotid glands from WKY subjected to sympathetic stimulation, with or without previous propranolol blockade. Diabetes and hypertension decreased the salivary secretion and increased SGLT1 expression in the luminal membrane of ductal cells, and their association exacerbated the regulations observed. After 30 min of sympathetic stimulation, SGLT1 increased in the luminal membrane of ductal cells, and that was blocked by previous injection of propranolol. CONCLUSIONS: SGLT1 expression increases in the luminal membrane of salivary gland ductal cells and the salivary flow decreases in diabetic and hypertensive rats, which may be related to sympathetic activity. This study highlights the water transporter role of SGLT1 in salivary glands, which, by increasing ductal water reabsorption, may explain the hyposalivation of diabetic and hypertensive subjects.

14.
Cardiovasc Diabetol ; 11: 100, 2012 Aug 16.
Article in English | MEDLINE | ID: mdl-22897936

ABSTRACT

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.


Subject(s)
Glucose Transporter Type 4/metabolism , Inflammation Mediators/blood , Insulin Resistance , Metabolic Syndrome/metabolism , Adiponectin/blood , Adipose Tissue, White/metabolism , Animals , Animals, Newborn , Biomarkers/blood , Blood Pressure , C-Reactive Protein/metabolism , Disease Models, Animal , Down-Regulation , Hypertension/blood , Hypertension/complications , Hypertension/physiopathology , Interleukin-6/blood , Lipids/blood , Male , Metabolic Syndrome/blood , Metabolic Syndrome/chemically induced , Metabolic Syndrome/immunology , Metabolic Syndrome/physiopathology , Muscle, Skeletal/metabolism , Myocardium/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Sodium Glutamate , Time Factors , Tumor Necrosis Factor-alpha/blood
15.
Eur J Pharmacol ; 689(1-3): 285-93, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22713545

ABSTRACT

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.


Subject(s)
Inflammation Mediators/antagonists & inhibitors , Insulin/physiology , Muscle Fibers, Skeletal/drug effects , Muscle, Skeletal/drug effects , Quercetin/pharmacology , Animals , Antioxidants/pharmacology , Down-Regulation/drug effects , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/prevention & control , Inflammation Mediators/metabolism , Male , Mice , Mice, Obese , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Quercetin/therapeutic use , Up-Regulation/drug effects
16.
J Endocrinol ; 211(1): 55-64, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21746792

ABSTRACT

Insulin replacement is the only effective therapy to manage hyperglycemia in type 1 diabetes mellitus (T1DM). Nevertheless, intensive insulin therapy has inadvertently led to insulin resistance. This study investigates mechanisms involved in the insulin resistance induced by hyperinsulinization. Wistar rats were rendered diabetic by alloxan injection, and 2 weeks later received saline or different doses of neutral protamine Hagedorn insulin (1.5, 3, 6, and 9 U/day) over 7 days. Insulinopenic-untreated rats and 6U- and 9U-treated rats developed insulin resistance, whereas 3U-treated rats revealed the highest grade of insulin sensitivity, but did not achieve good glycemic control as 6U- and 9U-treated rats did. This insulin sensitivity profile was in agreement with glucose transporter 4 expression and translocation in skeletal muscle, and insulin signaling, phosphoenolpyruvate carboxykinase/glucose-6-phosphatase expression and glycogen storage in the liver. Under the expectation that insulin resistance develops in hyperinsulinized diabetic patients, we believe insulin sensitizer approaches should be considered in treating T1DM.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Glucose/metabolism , Insulin Resistance/physiology , Insulin/therapeutic use , Liver/metabolism , Muscle, Skeletal/metabolism , Alloxan/adverse effects , Animals , Diabetes Mellitus, Experimental/chemically induced , Disease Models, Animal , Dose-Response Relationship, Drug , Forkhead Transcription Factors/metabolism , Glucose Transporter Type 4/metabolism , Glucose-6-Phosphatase/metabolism , Glycogen/metabolism , Hypoglycemic Agents/therapeutic use , Male , Nerve Tissue Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Wistar
17.
Cardiovasc Diabetol ; 10: 33, 2011 Apr 17.
Article in English | MEDLINE | ID: mdl-21496329

ABSTRACT

BACKGROUND: The effects of renal denervation on cardiovascular reflexes and markers of nephropathy in diabetic-hypertensive rats have not yet been explored. AIM: To evaluate the effects of renal denervation on nephropathy development mechanisms (blood pressure, cardiovascular autonomic changes, renal GLUT2) in diabetic-hypertensive rats. Forty-one male spontaneously hypertensive rats (SHR) ~250 g were injected with STZ or not; 30 days later, surgical renal denervation (RD) or sham procedure was performed; 15 days later, glycemia and albuminuria (ELISA) were evaluated. Catheters were implanted into the femoral artery to evaluate arterial pressure (AP) and heart rate variability (spectral analysis) one day later in conscious animals. Animals were killed, kidneys removed, and cortical renal GLUT2 quantified (Western blotting). RESULTS: Higher glycemia (p < 0.05) and lower mean AP were observed in diabetics vs. nondiabetics (p < 0.05). Heart rate was higher in renal-denervated hypertensive and lower in diabetic-hypertensive rats (384.8 +/- 37, 431.3+/- 36, 316.2 +/- 5, 363.8 +/- 12 bpm in SHR, RD-SHR,STZ-SHR and RD-STZ-SHR, respectively). Heart rate variability was higher in renal-denervated diabetic hypertensive rats (69.84 ± 37.91, 55.75 ± 25.21, 73.40 ±53.30, 148.4 ± 93 in SHR, RD-SHR, STZ-SHR- and RDSTZ-SHR, respectively, p < 0.05), as well as the LF component of AP variability (5.17 ± 5.24, 1.62 ± 0.9, 2.12 ±0.9, 7.38 ± 6.5 in SHR, RD-SHR, STZ-SHR and RDSTZ-SHR, respectively, p < 0.05). GLUT2 renal content was higher in all groups vs. SHR [corrected]. CONCLUSIONS: Renal denervation in diabetic-hypertensive rats improved previously reduced heart rate variability. The GLUT2 equally overexpressed by diabetes and renal denervation may represent a maximal derangement effect of each condition.


Subject(s)
Autonomic Denervation , Autonomic Nervous System/physiopathology , Diabetes Mellitus, Experimental/complications , Diabetic Nephropathies/etiology , Hypertension/complications , Kidney/innervation , Albuminuria/etiology , Albuminuria/physiopathology , Analysis of Variance , Animals , Blood Glucose/metabolism , Blood Pressure , Blotting, Western , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/physiopathology , Disease Models, Animal , Glucose Transporter Type 2/metabolism , Heart Rate , Hypertension/metabolism , Hypertension/physiopathology , Kidney/metabolism , Male , Rats , Rats, Inbred SHR , Reflex , Time Factors
18.
Am J Physiol Endocrinol Metab ; 299(6): E1028-37, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20841505

ABSTRACT

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.


Subject(s)
Cell Membrane/metabolism , Diabetes Mellitus/metabolism , Hypertension/metabolism , Parotid Gland/metabolism , Sodium-Glucose Transporter 1/metabolism , Submandibular Gland/metabolism , Sympathetic Nervous System/metabolism , Analysis of Variance , Animals , Blotting, Western , Cyclic AMP-Dependent Protein Kinases/metabolism , Diabetes Mellitus/physiopathology , Hypertension/physiopathology , Immunohistochemistry , Male , Parotid Gland/physiopathology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Signal Transduction , Submandibular Gland/physiopathology , Sympathetic Nervous System/physiopathology
19.
Cell Biochem Funct ; 28(2): 114-21, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20087847

ABSTRACT

Free fatty acids are known for playing a crucial role in the development of insulin resistance. High fat intake is known for impairing insulin sensitivity; however, the effect of vegetable-oil injections have never been investigated. The present study investigated the effects of daily subcutaneous injections (100 microL) of soybean (SB) and sunflower (SF) oils, during 7 days. Both treated groups developed insulin resistance as assessed by insulin tolerance test. The mechanism underlying the SB- and SF-induced insulin resistance was shown to involve GLUT4. In SB- and SF-treated animals, the GLUT4 protein expression was reduced approximately 20% and 10 min after an acute in vivo stimulus with insulin, the plasma membrane GLUT4 content was approximately 60% lower in white adipose tissue (WAT). No effects were observed in skeletal muscle. Additionally, both oil treatments increased mainly the content of palmitic acid ( approximately 150%) in WAT, which can contribute to explain the GLUT4 regulations. Altogether, the present study collects evidence that those oil treatments might generate insulin resistance by targeting GLUT4 expression and translocation specifically in WAT. These alterations are likely to be caused due to the specific local increase in saturated fatty acids that occurred as a consequence of oil daily injections.


Subject(s)
Adipose Tissue, White/metabolism , Diabetes Mellitus, Type 2/metabolism , Glucose Transporter Type 4/metabolism , Insulin Resistance , Animals , Cell Membrane/metabolism , Diabetes Mellitus, Type 2/chemically induced , Fatty Acids/blood , Injections, Subcutaneous , Insulin/metabolism , Male , Muscle, Skeletal/metabolism , Plant Oils/toxicity , Rats , Rats, Wistar , Soybean Oil/toxicity , Sunflower Oil
20.
J Endocrinol ; 198(1): 51-60, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18451064

ABSTRACT

Obesity and insulin resistance are highly correlated with metabolic disturbances. Both the excess and lack of adipose tissue can lead to severe insulin resistance and diabetes. Adipose tissue plays an active role in energy homeostasis, hormone secretion, and other proteins that affect insulin sensitivity, appetite, energy balance, and lipid metabolism. Rats with streptozotocin-induced diabetes during the neonatal period develop the classic diabetic picture of hyperglycemia, hypoinsulinemia, and insulin resistance in adulthood. Low body weight and reduced epididymal (EP) fat mass were also seen in this model. The aim of this study was to investigate the glucose homeostasis and metabolic repercussions on the adipose tissue following chronic treatment with antidiabetic drugs in these animals. In the 4th week post birth, diabetic animals started an 8-week treatment with pioglitazone, metformin, or insulin. Animals were then killed, EP fat pads were excised, and blood samples were collected for biological and biochemical assays. Pioglitazone and insulin treatments, but not metformin, reduced hyperglycemia, polydipsia, and polyphagia. Although all antidiabetic therapies improved insulin sensitivity, this was particularly noteworthy in the pioglitazone-treated rats. Furthermore, a recovery of adipose mass and insulin levels were observed in pioglitazone- and insulin-, but not metformin-treated animals. Treatments with insulin or pioglitazone were able to correct significantly, but not completely, the metabolic abnormalities, parallel to full recovery of adipose mass, indicating that not only the low insulin levels but also the lack of adipose tissue might play a significant role on the pathophysiology of this particular diabetes model.


Subject(s)
Adipose Tissue/metabolism , Diabetes Mellitus, Experimental/metabolism , Insulin Resistance , Animals , C-Peptide/analysis , Fatty Acids, Nonesterified/blood , Glucose/metabolism , Glucose Tolerance Test , Glucose Transporter Type 4/analysis , Glucose Transporter Type 4/genetics , Glycerides/blood , Lipolysis/drug effects , Male , Pioglitazone , RNA, Messenger/analysis , Rats , Rats, Wistar , Streptozocin , Thiazolidinediones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...