Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Nutrients ; 12(10)2020 Oct 13.
Article in English | MEDLINE | ID: mdl-33065982

ABSTRACT

In mammalian small intestine, glucose is primarily absorbed via Na-dependent glucose co-transporter (SGLT1) on the brush border membrane (BBM) of absorptive villus cells. Malabsorption of nutrients (e.g., glucose) leads to malnutrition, a common symptom of inflammatory bowel disease (IBD), where the mucosa is characterized by chronic inflammation. Inducible nitric oxide (iNO) is known to be elevated in IBD mucosa. SAMP1/YitFc (SAMP1) mouse is a spontaneous model of chronic ileitis that develops lesions in its terminal ileum, very similar to human IBD. How SGLT1 may be affected in SAMP1 model of chronic ileitis is unknown. Ten-week-old SAMP1 mice with AKR mice as control were treated with N6-(1-iminoethyl)-L-lysine dihydrochloride (L-NIL) to inhibit iNO production. Intracellular NO levels were found to be increased in villus cells from SAMP1 mice. Moreover, SGLT1 and Na+/K+-ATPase activities and BBM SGLT1 expression were significantly decreased. However, L-NIL treatment reduced the intracellular iNO production, and reversed both downregulated SGLT1 and Na+/K+-ATPase activities in SAMP1 mice. Inhibition of iNO by L-NIL treatment also significantly reversed the BBM SGLT1 protein expression in SAMP1 mice. L-NIL reversed the inflammation mediated downregulation of SGLT1 activity by restoring the BBM SGLT1 expression. Thus, regulation of SGLT1 in chronic ileitis is likely mediated by iNO.


Subject(s)
Biological Transport/drug effects , Crohn Disease/metabolism , Glucose/metabolism , Ileum/metabolism , Nitric Oxide/physiology , Sodium/metabolism , Animals , Chronic Disease , Disease Models, Animal , Gene Expression , Membrane Proteins , Mice, Transgenic , Microvilli/metabolism , Nitric Oxide/metabolism , Nuclear Proteins , Sodium-Glucose Transporter 1/genetics , Sodium-Glucose Transporter 1/metabolism , Sodium-Glucose Transporter 1/physiology , Sodium-Potassium-Exchanging ATPase/metabolism
2.
Biochim Biophys Acta Biomembr ; 1861(10): 182985, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31082355

ABSTRACT

In small intestine, sodium-glucose cotransporter SGLT1 provides the main mechanism for sugar uptake. We investigated the effect of membrane phospholipids (PL) on this transport in rabbit ileal brush border membrane vesicles (BBMV). For this, PL of different charge, length, and saturation were incorporated into BBMV. Transport was measured related to (i) membrane surface charge (membrane-bound MC540 fluorescence), (ii) membrane thickness (PL incorporation of different acyl chain length), and (iii) membrane fluidity (r12AS, fluorescence anisotropy of 12-AS). Compared to phosphatidylcholine (PC) carrying a neutral head group, inhibition of SGLT1 increased considerably with the acidic phosphatidic acid (PA) and phosphatidylinositol (PI) that increase membrane negative surface charge. The order of PL potency was PI>PA > PE = PS > PC. Inhibition by acidic PA-oleate was 5-times more effective than with neutral PE (phosphatidylethanolamine)-oleate. Lineweaver-Burk plot indicated uncompetitive inhibition of SGLT1 by PA. When membrane thickness was increased by neutral PC of varying acyl chain length, transport was increasingly inhibited by 16:1 PC to 22:1 PC. Even more pronounced inhibition was observed with mono-unsaturated instead of saturated acyl chains which increased membrane fluidity (indicated by decreased r12AS). In conclusion, sodium-dependent glucose transport of rabbit ileal BBMV is modulated by (i) altered membrane surface charge, (ii) length of acyl chains via membrane thickness, and (iii) saturation of PL acyl chains altering membrane fluidity. Transport was attenuated by charged PL with longer and unsaturated acyl residues. Alterations of PL may provide a principle for attenuating dietary glucose uptake.


Subject(s)
Glucose Transport Proteins, Facilitative/metabolism , Membrane Fluidity/drug effects , Sodium-Glucose Transporter 1/metabolism , Animals , Biological Transport , Fatty Acids/metabolism , Fluorescence Polarization/methods , Glucose/metabolism , Glucose Transport Proteins, Facilitative/physiology , Ileum/metabolism , Intestine, Small/metabolism , Male , Microvilli/metabolism , Microvilli/physiology , Phosphatidic Acids/chemistry , Phosphatidylcholines/chemistry , Phosphatidylinositols/chemistry , Phospholipids/metabolism , Phospholipids/physiology , Rabbits , Sodium/metabolism , Sodium-Glucose Transporter 1/physiology , Transport Vesicles/metabolism , Transport Vesicles/physiology
3.
Physiol Rep ; 7(9): e14090, 2019 05.
Article in English | MEDLINE | ID: mdl-31062524

ABSTRACT

Kinetic characterization of electrogenic sodium-dependent transport in Ussing chambers of d-glucose and d-galactose demonstrated sigmoidal/Hill kinetics in the porcine jejunum and ileum, with the absence of transport in the distal colon. In the jejunum, a high-affinity, super-low-capacity (Ha/sLc) kinetic system accounted for glucose transport, and a low-affinity, low-capacity (La/Lc) kinetic system accounted for galactose transport. In contrast, the ileum demonstrated a high-affinity, super-high-capacity (Ha/sHc) glucose transport and a low-affinity, high-capacity (La/Hc) galactose transport systems. Jejunal glucose transport was not inhibited by dapagliflozin, but galactose transport was inhibited. Comparatively, ileal glucose and galactose transport were both sensitive to dapagliflozin. Genomic and gene expression analyses identified 10 of the 12 known SLC5A family members in the porcine jejunum, ileum, and distal colon. Dominant SGLT1 (SLC5A1) and SGLT3 (SLC5A4) expression was associated with the sigmoidal Ha/sLc glucose and La/Lc galactose transport systems in the jejunum. Comparatively, the dominant expression of SGLT1 (SLC5A1) in the ileum was only associated with Ha glucose and La galactose kinetic systems. However, the sigmoidal kinetics and overall high capacity (Hc) of transport is unlikely accounted for by SGLT1 (SLC5A1) alone. Finally, the absence of transport and lack of pharmacological inhibition in the colon was associated with the poor expression of SLC5A genes. Altogether, the results demonstrated intestinal segregation of monosaccharide transport fit different sigmoidal kinetic systems. This reveals multiple transporter populations in each system, supported by gene expression profiles and pharmacological inhibition. Overall, this work demonstrates a complexity to transporter involvement in intestinal electrogenic monosaccharide absorption systems not previously defined.


Subject(s)
Intestinal Mucosa/metabolism , Membrane Transport Proteins/physiology , Monosaccharides/metabolism , Sus scrofa/metabolism , Animals , Biological Transport/physiology , Colon/metabolism , Galactose/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation/physiology , Glucose/metabolism , Ileum/metabolism , Intestinal Absorption/physiology , Jejunum/metabolism , Sodium-Glucose Transport Proteins/biosynthesis , Sodium-Glucose Transport Proteins/genetics , Sodium-Glucose Transporter 1/physiology
4.
Diabetologia ; 61(10): 2087-2097, 2018 10.
Article in English | MEDLINE | ID: mdl-30132032

ABSTRACT

The concentration of glucose in plasma is held within narrow limits (4-10 mmol/l), primarily to ensure fuel supply to the brain. Kidneys play a role in glucose homeostasis in the body by ensuring that glucose is not lost in the urine. Three membrane proteins are responsible for glucose reabsorption from the glomerular filtrate in the proximal tubule: sodium-glucose cotransporters SGLT1 and SGLT2, in the apical membrane, and GLUT2, a uniporter in the basolateral membrane. 'Knockout' of these transporters in mice and men results in the excretion of filtered glucose in the urine. In humans, intravenous injection of the plant glucoside phlorizin also results in excretion of the full filtered glucose load. This outcome and the finding that, in an animal model, phlorizin reversed the symptoms of diabetes, has stimulated the development and successful introduction of SGLT2 inhibitors, gliflozins, in the treatment of type 2 diabetes mellitus. Here we summarise the current state of our knowledge about the physiology of renal glucose handling and provide background to the development of SGLT2 inhibitors for type 2 diabetes treatment.


Subject(s)
Glucose Transporter Type 2/physiology , Glucose/metabolism , Kidney/metabolism , Sodium-Glucose Transporter 1/physiology , Sodium-Glucose Transporter 2/physiology , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/metabolism , Drug Design , Glucose Transporter Type 2/genetics , Glycosuria/metabolism , HEK293 Cells , Homeostasis , Humans , Hypoglycemic Agents/pharmacology , Kidney Tubules/metabolism , Kidney Tubules, Proximal/metabolism , Mice , Mice, Knockout , Phlorhizin/pharmacology , Sodium-Glucose Transporter 1/genetics , Sodium-Glucose Transporter 2/genetics , Sodium-Glucose Transporter 2 Inhibitors/pharmacology
5.
Yakugaku Zasshi ; 138(7): 939-943, 2018.
Article in Japanese | MEDLINE | ID: mdl-29962473

ABSTRACT

 It is well-known that metabolic remodeling occurs in the presence of cardiomyopathy induced by cardiac ischemia and hypertrophy, and diabetes mellitus. It is also known that a novel cardiac glucose transporter, sodium-glucose co-transporter 1 (SGLT1), is expressed in the human heart. However, the role of SGLT1 in the development of cardiac metabolic remodeling is still unclear. Recent studies demonstrated that SGLT1 activation improves ischemia-reperfusion-induced cardiac injury, and increased SGLT1 gene expression is observed in hypertrophic, ischemic, and diabetic cardiomyopathy in human hearts. Moreover, increases in SGLT1 protein expression cause cardiac remodeling such as hypertrophy and increased interstitial fibrosis in mice. We demonstrated that ischemia-reperfusion-induced cardiac injury was potentiated in SGLT1-deficient mice. In contrast, chronic pressure overload induced by transverse aortic constriction (TAC) caused cardiac hypertrophy and reduced left ventricular fractional shortening in C57BL/6J wild-type mice. Moreover, the TAC-induced hypertrophied heart showed increased SGLT1 and AMPKαprotein expressions. These results suggest the different effects of SGLT1 activation on cardiac diseases such as acute ischemia-reperfusion-induced cardiac injury and chronically-induced cardiac hypertrophy. Thus, SGLT1 may be a novel therapeutic target for the treatment of patients with cardiac diseases such as ischemic and hypertrophic cardiomyopathy.


Subject(s)
Sodium-Glucose Transporter 1/physiology , Ventricular Remodeling/genetics , Animals , Cardiomegaly/complications , Cardiomegaly/genetics , Cardiomyopathy, Hypertrophic/drug therapy , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/metabolism , Diabetes Complications/genetics , Gene Expression , Humans , Molecular Targeted Therapy , Myocardial Ischemia/complications , Myocardial Ischemia/drug therapy , Myocardial Ischemia/genetics , Myocardium/metabolism , Sodium-Glucose Transporter 1/genetics , Sodium-Glucose Transporter 1/metabolism
6.
Nat Rev Nephrol ; 13(1): 11-26, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27941935

ABSTRACT

The kidney has a pivotal role in maintaining glucose homeostasis by using glucose as a metabolic fuel, by producing glucose through gluconeogenesis, and by reabsorbing all filtered glucose through the sodium-glucose cotransporters SGLT1 and SGLT2 located in the proximal tubule. In patients with diabetes, the maximum glucose reabsorptive capacity (TmG) of the kidney, as well as the threshold for glucose spillage into the urine, are elevated, contributing to the pathogenesis of hyperglycaemia. By reducing the TmG and, more importantly, the threshold of glucosuria, SGLT2 inhibitors enhance glucose excretion, leading to a reduction in fasting and postprandial plasma glucose levels and improvements in both insulin secretion and insulin sensitivity. The beneficial effects of SGLT2 inhibition extend beyond glycaemic control, however, with new studies demonstrating that inhibition of renal glucose reabsorption reduces blood pressure, ameliorates glucotoxicity and induces haemodynamic effects that lead to improved cardiovascular and renal outcomes in patients with type 2 diabetes mellitus. In this Review we examine the role of SGLT2 and SGLT1 in the regulation of renal glucose reabsorption in health and disease and the effect of SGLT2 inhibition on renal function, glucose homeostasis, and cardiovascular disease.


Subject(s)
Cardiovascular Diseases/etiology , Glucose/metabolism , Homeostasis , Kidney/metabolism , Sodium-Glucose Transporter 2 Inhibitors , Animals , Blood Glucose , Cardiovascular Diseases/prevention & control , Humans , Renal Reabsorption , Sodium-Glucose Transporter 1/physiology , Sodium-Glucose Transporter 2/physiology
7.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 45(6): 598-606, 2016 05 25.
Article in Chinese | MEDLINE | ID: mdl-28247603

ABSTRACT

Objective: To investigate the role of glucose transporter 1 (GLUT1) and sodium-glucose cotransporter 1 (SGLT1) in high glucose dialysate-induced peritoneal fibrosis. Methods: Thirty six male SD rats were randomly divided into 6 groups (6 in each):normal control group, sham operation group, peritoneal dialysis group (PD group), PD+phloretin group (PD+T group), PD+phlorizin group (PD+Z group), PD+phloretin+phlorizin group (PD+T+Z group). Rat model of uraemia was established using 5/6 nephrotomy, and 2.5% dextrose peritoneal dialysis solution was used in peritoneal dialysis. Peritoneal equilibration test was performed 24 h after dialysis to evaluate transport function of peritoneum in rats; HE staining was used to observe the morphology of peritoneal tissue; and immunohistochemistry was used to detect the expression of GLUT1, SGLT1, TGF-ß1 and connective tissue growth factor (CTGF) in peritoneum. Human peritoneal microvascular endothelial cells (HPECs) were divided into 5 groups:normal control group, peritoneal dialysis group (PD group), PD+phloretin group (PD+T group), PD+phlorezin group (PD+Z group), and PD+phloretin+phlorezin group (PD+T+Z group). Real time PCR and Western blotting were used to detect mRNA and protein expressions of GLUT1, SGLT1, TGF-ß1, CTGF in peritoneal membrane and HPECs. Results:In vivo, compared with sham operation group, rats in PD group had thickened peritoneum, higher ultrafiltration volume, and the mRNA and protein expressions of GLUT1, SGLT1, CTGF, TGF-ß1 were significantly increased (all P<0.05); compared with PD group, thickened peritoneum was attenuated, and the mRNA and protein expressions of GLUT1, SGLT1, CTGF, TGF-ß1 were significantly decreased in PD+T, PD+Z and PD+T+Z groups (all P<0.05). Pearson's correlation analysis showed that the expressions of GLUT1, SGLT1 in peritoneum were positively correlated with the expressions of TGF-ß1 and CTGF (all P<0.05). In vitro, the mRNA and protein expressions of GLUT1, SGLT1, TGF-ß1, CTGF were significantly increased in HPECs of peritoneal dialysis group (all P<0.05), and those in PD+T, PD+Z, and PD+T+Z groups were decreased (all P<0.05). Pearson's correlation analysis showed that the expressions of GLUT1, SGLT1 in HPECs were positively correlated with the expressions of TGF-ß1 and CTGF (all P<0.05). Conclusion: High glucose peritoneal dialysis fluid may promote peritoneal fibrosis by upregulating the expressions of GLUT1 and SGLT1.


Subject(s)
Dialysis Solutions/adverse effects , Dialysis Solutions/pharmacology , Glucose Transporter Type 1/drug effects , Glucose Transporter Type 1/physiology , Glucose/adverse effects , Glucose/pharmacology , Hemodiafiltration/adverse effects , Hemodiafiltration/methods , Peritoneal Dialysis/adverse effects , Peritoneal Dialysis/methods , Peritoneal Fibrosis/chemically induced , Peritoneal Fibrosis/genetics , Peritoneum/chemistry , Peritoneum/drug effects , Peritoneum/pathology , Sodium-Glucose Transporter 1/drug effects , Sodium-Glucose Transporter 1/physiology , Uremia/chemically induced , Animals , Cells, Cultured , Connective Tissue Growth Factor/analysis , Connective Tissue Growth Factor/drug effects , Dialysis Solutions/chemistry , Gene Expression Regulation/drug effects , Glucose Transporter Type 1/analysis , Humans , Male , Peritoneal Fibrosis/physiopathology , Phloretin , Phlorhizin , RNA, Messenger , Rats , Rats, Sprague-Dawley , Sodium-Glucose Transporter 1/analysis , Transforming Growth Factor beta1/analysis , Transforming Growth Factor beta1/drug effects
8.
Drug Metab Dispos ; 44(3): 283-96, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26658676

ABSTRACT

Hydrolysis by lactase-phloridzin hydrolase (LPH) is the first and critical step in the absorption of isoflavonoid glucosides. However, the absorption characteristics of calycosin-7-O-ß-d-glucoside (CG) slightly differ from other isoflavonoid glucosides. In this study, we used the rat intestinal perfusion model and performed pharmacokinetic studies and in vitro experiments to determine the factors influencing CG absorption and disposition. After oral administration of isoflavonoid glucosides, LPH was found to play minimal or no role on the hydrolysis of CG, in contrast to that of daidzin. CG was mainly transported into the small intestinal cells by sodium-dependent glucose transporter 1 (SGLT-1) as intact. This pathway could be the main mechanism underlying the high permeability of CG in the small intestine. CG was likely to be hydrolyzed in enterocytes to its aglycone calycosin by broad-specific ß-glucuronides (BSßG) and glucocerebrosidase or rapidly metabolized. Calycosin was also rapidly and extensively metabolized to 3'-glucuronide in the enterocytes and liver, and the glucuronidation rates of calycosin and CG were much higher in the former. The metabolites were also transported into lumen by breast cancer resistance protein and multidrug resistance-associated protein 2. In conclusion, the enterocytes could be an important site for CG absorption, deglycosylation, and metabolism in rats. This study could contribute to the theoretical foundation and mechanism of absorption and disposition of flavonoid compounds.


Subject(s)
Glucosides/metabolism , Intestine, Small/metabolism , Isoflavones/metabolism , Protein Transport/physiology , Sodium-Glucose Transporter 1/physiology , Animals , Caco-2 Cells , Cell Line, Tumor , Enterocytes/metabolism , Glucosylceramidase/metabolism , Glucuronides/metabolism , Glycosylation , Humans , Intestinal Absorption/physiology , Male , Permeability , Rats , Rats, Sprague-Dawley
9.
Med Clin (Barc) ; 147 Suppl 1: 26-29, 2016 Nov.
Article in English, Spanish | MEDLINE | ID: mdl-28760222

ABSTRACT

Selectivity is the property of a drug to preferentially bind to a biological structure. Most drugs can bind and stimulate or inhibit more than one system. Therefore, it is important that they are selective for the intended site and that the doses used do not have effects on other sites, which could provoke adverse reactions. Selectivity is assessed through in vitro experiments on organs or isolated cells. If the aim is to compare drugs, the experiment should be conducted in the same tissue and with the same design. Even so, the results cannot be directly extrapolated to clinical practice due to the influence of pharmacokinetic properties, which allow an adequate dose of the drug to reach the target site. Sodium-glucose cotransporter-2 inhibitors (SGLT2i) are able to inhibit renal SGLT2 without modifying intestinal SGLT1, whose inhibition could produce gastrointestinal adverse reactions. The concentration needed to inhibit each of the transporters is calculated, as well as the ratio between the concentration that inhibits SGLT1 and the concentration needed to inhibit SGLT2. The higher the ratio, the greater the selectivity and the lower the risk of gastrointestinal adverse reactions. The three SGLT2i recently introduced in the therapeutic arsenal are sufficiently selective for SGLT2 to make effects on intestinal SGLT1 unlikely. To differentiate the components of this therapeutic class, its pharmacokinetic properties should be analysed rather than its pharmacodynamic characteristics, such as selectivity.


Subject(s)
Hypoglycemic Agents/pharmacology , Sodium-Glucose Transporter 1/antagonists & inhibitors , Sodium-Glucose Transporter 2 Inhibitors , Diabetes Mellitus, Type 2/drug therapy , Glucose , Humans , Sodium , Sodium-Glucose Transporter 1/physiology , Sodium-Glucose Transporter 2/physiology
10.
J Endocrinol ; 224(3): 205-14, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25486965

ABSTRACT

Oral ingestion of carbohydrate triggers glucagon-like peptide 1 (GLP1) secretion, but the molecular mechanism remains elusive. By measuring GLP1 concentrations in murine portal vein, we found that the ATP-sensitive K(+) (KATP) channel is not essential for glucose-induced GLP1 secretion from enteroendocrine L cells, while the sodium-glucose co-transporter 1 (SGLT1) is required, at least in the early phase (5 min) of secretion. By contrast, co-administration of the α-glucosidase inhibitor (α-GI) miglitol plus maltose evoked late-phase secretion in a glucose transporter 2-dependent manner. We found that GLP1 secretion induced by miglitol plus maltose was significantly higher than that by another α-GI, acarbose, plus maltose, despite the fact that acarbose inhibits maltase more potently than miglitol. As miglitol activates SGLT3, we compared the effects of miglitol on GLP1 secretion with those of acarbose, which failed to depolarize the Xenopus laevis oocytes expressing human SGLT3. Oral administration of miglitol activated duodenal enterochromaffin (EC) cells as assessed by immunostaining of phosphorylated calcium-calmodulin kinase 2 (phospho-CaMK2). In contrast, acarbose activated much fewer enteroendocrine cells, having only modest phospho-CaMK2 immunoreactivity. Single administration of miglitol triggered no GLP1 secretion, and GLP1 secretion by miglitol plus maltose was significantly attenuated by atropine pretreatment, suggesting regulation via vagal nerve. Thus, while α-GIs generally delay carbohydrate absorption and potentiate GLP1 secretion, miglitol also activates duodenal EC cells, possibly via SGLT3, and potentiates GLP1 secretion through the parasympathetic nervous system.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Enteroendocrine Cells/drug effects , Glucagon-Like Peptide 1/metabolism , Glycoside Hydrolase Inhibitors/pharmacology , Sodium-Glucose Transport Proteins/metabolism , 1-Deoxynojirimycin/pharmacology , Acarbose/pharmacology , Animals , Enteroendocrine Cells/metabolism , Glucose Transporter Type 2/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sodium-Glucose Transport Proteins/genetics , Sodium-Glucose Transporter 1/physiology , Xenopus laevis
11.
J Endocrinol ; 222(2): 191-200, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24891433

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP), a gut hormone secreted from intestinal K-cells, potentiates insulin secretion. Both K-cells and pancreatic ß-cells are glucose-responsive and equipped with a similar glucose-sensing apparatus that includes glucokinase and an ATP-sensitive K(+) (KATP) channel comprising KIR6.2 and sulfonylurea receptor 1. In absorptive epithelial cells and enteroendocrine cells, sodium glucose co-transporter 1 (SGLT1) is also known to play an important role in glucose absorption and glucose-induced incretin secretion. However, the glucose-sensing mechanism in K-cells is not fully understood. In this study, we examined the involvement of SGLT1 (SLC5A1) and the KATP channels in glucose sensing in GIP secretion in both normal and streptozotocin-induced diabetic mice. Glimepiride, a sulfonylurea, did not induce GIP secretion and pretreatment with diazoxide, a KATP channel activator, did not affect glucose-induced GIP secretion in the normal state. In mice lacking KATP channels (Kir6.2(-/-) mice), glucose-induced GIP secretion was enhanced compared with control (Kir6.2(+) (/) (+)) mice, but was completely blocked by the SGLT1 inhibitor phlorizin. In Kir6.2(-/-) mice, intestinal glucose absorption through SGLT1 was enhanced compared with that in Kir6.2(+) (/) (+) mice. On the other hand, glucose-induced GIP secretion was enhanced in the diabetic state in Kir6.2(+) (/) (+) mice. This GIP secretion was partially blocked by phlorizin, but was completely blocked by pretreatment with diazoxide in addition to phlorizin administration. These results demonstrate that glucose-induced GIP secretion depends primarily on SGLT1 in the normal state, whereas the KATP channel as well as SGLT1 is involved in GIP secretion in the diabetic state in vivo.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Gastric Inhibitory Polypeptide/metabolism , KATP Channels/physiology , Sodium-Glucose Transporter 1/physiology , Animals , Diazoxide/pharmacology , Glucose/pharmacology , Mice , Phlorhizin/pharmacology , Potassium Channels, Inwardly Rectifying/deficiency , Potassium Channels, Inwardly Rectifying/physiology , Sulfonylurea Compounds/pharmacology
12.
Crit Care Med ; 42(1): 57-65, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23963126

ABSTRACT

OBJECTIVES: Providing effective enteral nutrition is important during critical illness. In health, glucose is absorbed from the small intestine via sodium-dependent glucose transporter-1 and glucose transporter-2, which may both be regulated by intestinal sweet taste receptors. We evaluated the effect of critical illness on glucose absorption and expression of intestinal sodium-dependent glucose transporter-1, glucose transporter-2, and sweet taste receptors in humans and mice. DESIGN: Prospective observational study in humans and mice. SETTING: ICU and university-affiliated research laboratory. SUBJECTS: Human subjects were 12 critically ill patients and 12 healthy controls. In the laboratory 16-week-old mice were studied. INTERVENTIONS: Human subjects underwent endoscopy. Glucose (30 g) and 3-O-methylglucose (3 g), used to estimate glucose absorption, were infused intraduodenally over 30 minutes. Duodenal mucosa was biopsied before and after infusion. Mice were randomized to cecal ligation and puncture to model critical illness (n = 16) or sham laparotomy (control) (n = 8). At day 5, mice received glucose (100 mg) and 3-O-methylglucose (10 mg) infused intraduodenally prior to mucosal tissue collection. MEASUREMENTS AND MAIN RESULTS: Quantitative polymerase chain reaction was performed to measure absolute (human) and relative levels of sodium-dependent glucose transporter-1, glucose transporter-2, and taste receptor type 1 member 2 (T1R2) transcripts. Blood samples were assayed for 3-O-methylglucose to estimate glucose absorption. Glucose absorption was three-fold lower in critically ill humans than in controls (p = 0.002) and reduced by a similar proportion in cecal ligation and puncture mice (p = 0.004). In critically ill patients, duodenal levels of sodium-dependent glucose transporter-1, glucose transporter-2, and T1R2 transcript were reduced 49% (p < 0.001), 50% (p = 0.009), and 85% (p = 0.007), whereas in the jejunum of cecal ligation and puncture mice sodium-dependent glucose transporter-1, glucose transporter-2, and T1R2 transcripts were reduced by 55% (p < 0.001), 50% (p = 0.002), and 69% (p = 0.004). CONCLUSIONS: Critical illness is characterized by markedly diminished glucose absorption, associated with reduced intestinal expression of glucose transporters (sodium-dependent glucose transporter-1 and glucose transporter-2) and sweet taste receptor transcripts. These changes are paralleled in cecal ligation and puncture mice.


Subject(s)
Critical Illness , Glucose/metabolism , Intestinal Absorption/physiology , Intestines/physiopathology , 3-O-Methylglucose/metabolism , Adult , Aged , Animals , Disease Models, Animal , Duodenum/physiopathology , Female , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 1/physiology , Glucose Transporter Type 2/metabolism , Glucose Transporter Type 2/physiology , Humans , Male , Mice , Middle Aged , Prospective Studies , Real-Time Polymerase Chain Reaction , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/physiology , Sodium-Glucose Transporter 1/metabolism , Sodium-Glucose Transporter 1/physiology , Young Adult
13.
Am J Physiol Renal Physiol ; 306(2): F188-93, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24226519

ABSTRACT

In the kidney, the sodium-glucose cotransporters SGLT2 and SGLT1 are thought to account for >90 and ∼3% of fractional glucose reabsorption (FGR), respectively. However, euglycemic humans treated with an SGLT2 inhibitor maintain an FGR of 40-50%, mimicking values in Sglt2 knockout mice. Here, we show that oral gavage with a selective SGLT2 inhibitor (SGLT2-I) dose dependently increased urinary glucose excretion (UGE) in wild-type (WT) mice. The dose-response curve was shifted leftward and the maximum response doubled in Sglt1 knockout (Sglt1-/-) mice. Treatment in diet with the SGLT2-I for 3 wk maintained 1.5- to 2-fold higher urine glucose/creatinine ratios in Sglt1-/- vs. WT mice, associated with a temporarily greater reduction in blood glucose in Sglt1-/- vs. WT after 24 h (-33 vs. -11%). Subsequent inulin clearance studies under anesthesia revealed free plasma concentrations of the SGLT2-I (corresponding to early proximal concentration) close to the reported IC50 for SGLT2 in mice, which were associated with FGR of 64 ± 2% in WT and 17 ± 2% in Sglt1-/-. Additional intraperitoneal application of the SGLT2-I (maximum effective dose in metabolic cages) increased free plasma concentrations ∼10-fold and reduced FGR to 44 ± 3% in WT and to -1 ± 3% in Sglt1-/-. The absence of renal glucose reabsorption was confirmed in male and female Sglt1/Sglt2 double knockout mice. In conclusion, SGLT2 and SGLT1 account for renal glucose reabsorption in euglycemia, with 97 and 3% being reabsorbed by SGLT2 and SGLT1, respectively. When SGLT2 is fully inhibited by SGLT2-I, the increase in SGLT1-mediated glucose reabsorption explains why only 50-60% of filtered glucose is excreted.


Subject(s)
Glucose/metabolism , Kidney/metabolism , Sodium-Glucose Transporter 1/physiology , Sodium-Glucose Transporter 2 Inhibitors , Sodium-Glucose Transporter 2/genetics , Animals , Benzhydryl Compounds/pharmacology , Biological Transport, Active/physiology , Blood Glucose/physiology , Dose-Response Relationship, Drug , Drinking/physiology , Eating/physiology , Female , Glucosides/pharmacology , Glycosuria/metabolism , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Male , Mice , Mice, Knockout , Sodium-Glucose Transporter 1/genetics , Urodynamics/drug effects , Urodynamics/genetics
15.
Vet Immunol Immunopathol ; 155(3): 171-81, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23886446

ABSTRACT

Interleukins (IL), aside from their role in the regulation of the immune cascade, they have also been shown to modulate intestinal transport function. IL-1ß is a potent inflammatory cytokine involved in many important cellular functions. The aim of this work was to study the in vitro effect of IL-1ß on d-galactose transport across intestinal epithelia in rabbit jejunum and Caco-2 cells. The results showed that d-galactose intestinal absorption was diminished in IL-1ß treated jejunum rabbits without affecting the Na(+), K(+)-ATPase activity. The presence of IL-1 cell-surface receptors was confirmed by addition to tissue of a specific IL-1 receptor antagonist (IL-1ra). The cytokine did not inhibit either the uptake of d-galactose nor modified the sodium-glucose transport (SGLT1) protein levels in the brush border membrane vesicles, suggesting an indirect IL effect. The IL-inhibition was significantly reversed in the presence of inhibitors of protein kinase C (PKC) and mitogen-activated protein kinases (MAPKs). The proteasome selective inhibitor completely abolished the IL-effect. Furthermore, the cytokine inhibition on galactose transport related to NF-kB activation was also confirmed in Caco-2 cells. In summary, the direct addition of IL-1ß to intestinal epithelia inhibits d-galactose transport by a possible reduction in the SGLT1 activity. This event may be mediated by several transduction pathways activated during the inflammatory processes related to several protein kinases and nuclear factor, NF-kB. The IL-effect is independent of hormonal milieu and nervous stimuli.


Subject(s)
Galactose/metabolism , Interleukin-1beta/pharmacology , Intestinal Mucosa/metabolism , NF-kappa B/physiology , Protein Kinase C/physiology , Animals , Biological Transport , Caco-2 Cells , Cyclic AMP-Dependent Protein Kinases/physiology , Humans , MAP Kinase Signaling System , Male , Rabbits , Sodium-Glucose Transporter 1/physiology
16.
Biochim Biophys Acta ; 1828(11): 2394-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23774524

ABSTRACT

The Na(+)-coupled glucose transporter SGLT1 (SLC5A1) accomplishes concentrative cellular glucose uptake even at low extracellular glucose concentrations. The carrier is expressed in renal proximal tubules, small intestine and a variety of nonpolarized cells including several tumor cells. The present study explored whether SGLT1 activity is regulated by caveolin-1, which is known to regulate the insertion of several ion channels and carriers in the cell membrane. To this end, SGLT1 was expressed in Xenopus oocytes with or without additional expression of caveolin-1 and electrogenic glucose transport determined by dual electrode voltage clamp experiments. In SGLT1-expressing oocytes, but not in oocytes injected with water or caveolin-1 alone, the addition of glucose to the extracellular bath generated an inward current (Ig), which was increased following coexpression of caveolin-1. Kinetic analysis revealed that caveolin-1 increased maximal Ig without significantly modifying the glucose concentration required to trigger half maximal Ig (KM). According to chemiluminescence and confocal microscopy, caveolin-1 increased SGLT1 protein abundance in the cell membrane. Inhibition of SGLT1 insertion by brefeldin A (5µM) resulted in a decline of Ig, which was similar in the absence and presence of caveolin-1. In conclusion, caveolin-1 up-regulates SGLT1 activity by increasing carrier protein abundance in the cell membrane, an effect presumably due to stimulation of carrier protein insertion into the cell membrane.


Subject(s)
Caveolin 1/physiology , Sodium-Glucose Transporter 1/physiology , Up-Regulation/physiology , Animals , Cell Membrane/metabolism , Kinetics , Sodium-Glucose Transporter 1/metabolism , Xenopus
17.
Radiother Oncol ; 107(2): 247-51, 2013 May.
Article in English | MEDLINE | ID: mdl-23602371

ABSTRACT

BACKGROUND AND PURPOSE: EGFR and the sodium-dependent glucose transporter, SGLT1, are found in complex after radiation treatment. The aim of this study was to elucidate the role of EGFR in glucose uptake and chromatin remodeling. MATERIAL AND METHODS: Glucose accumulation was quantified with help of (3)H-glucose. Involvement of SGLT was detected by a specific inhibitor. Role of EGFR was proved by EGFR overexpression and siRNA driven knockdown. Functional endpoints were intracellular ATP levels, protein expression, residual DNA-damage and colony formation. RESULTS: EGFR/SGLT1 interactions in response to ionizing radiation were associated with increased glucose uptake. Nevertheless, tumor cells exhibit ATP depletion following irradiation. Recovery from radiation-induced ATP crisis was EGFR/SGLT-dependent and associated with increased cell survival and improved DNA-repair. The blockage of either EGFR or SGLT inhibited ATP level recovery and histone H3 modifications crucial for both chromatin remodeling and DNA repair in response to irradiation. Inhibition of the acetyltransferase TIP60, which is essential for histone H3-K9 acetylation and ATM activation, prevented energy crisis and chromatin remodeling. CONCLUSIONS: Radiation-associated interactions between SGLT1 and EGFR resulted in increased glucose uptake, which counteracts the ATP crisis in tumor cells due to chromatin remodeling. The blockage of recovery from ATP crisis led to radio-sensitization in tumor cells.


Subject(s)
Chromatin Assembly and Disassembly/radiation effects , ErbB Receptors/physiology , Sodium-Glucose Transporter 1/physiology , Adenosine Triphosphate/analysis , Animals , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Glucose/metabolism , Humans , Phlorhizin/pharmacology
18.
J Pharmacol Exp Ther ; 345(2): 250-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23487174

ABSTRACT

LX4211 [(2S,3R,4R,5S,6R)-2-(4-chloro-3-(4-ethoxybenzyl)phenyl)-6-(methylthio)tetrahydro-2H-pyran-3,4,5-triol], a dual sodium/glucose cotransporter 1 (SGLT1) and SGLT2 inhibitor, is thought to decrease both renal glucose reabsorption by inhibiting SGLT2 and intestinal glucose absorption by inhibiting SGLT1. In clinical trials in patients with type 2 diabetes mellitus (T2DM), LX4211 treatment improved glycemic control while increasing circulating levels of glucagon-like peptide 1 (GLP-1) and peptide YY (PYY). To better understand how LX4211 increases GLP-1 and PYY levels, we challenged SGLT1 knockout (-/-) mice, SGLT2-/- mice, and LX4211-treated mice with oral glucose. LX4211-treated mice and SGLT1-/- mice had increased levels of plasma GLP-1, plasma PYY, and intestinal glucose during the 6 hours after a glucose-containing meal, as reflected by area under the curve (AUC) values, whereas SGLT2-/- mice showed no response. LX4211-treated mice and SGLT1-/- mice also had increased GLP-1 AUC values, decreased glucose-dependent insulinotropic polypeptide (GIP) AUC values, and decreased blood glucose excursions during the 6 hours after a challenge with oral glucose alone. However, GLP-1 and GIP levels were not increased in LX4211-treated mice and were decreased in SGLT1-/- mice, 5 minutes after oral glucose, consistent with studies linking decreased intestinal SGLT1 activity with reduced GLP-1 and GIP levels 5 minutes after oral glucose. These data suggest that LX4211 reduces intestinal glucose absorption by inhibiting SGLT1, resulting in net increases in GLP-1 and PYY release and decreases in GIP release and blood glucose excursions. The ability to inhibit both intestinal SGLT1 and renal SGLT2 provides LX4211 with a novel dual mechanism of action for improving glycemic control in patients with T2DM.


Subject(s)
Glucagon-Like Peptide 1/blood , Glucose/metabolism , Glycosides/pharmacology , Hypoglycemic Agents/pharmacology , Intestinal Absorption/drug effects , Peptide YY/blood , Sodium-Glucose Transporter 1/antagonists & inhibitors , Sodium-Glucose Transporter 2 Inhibitors , Animals , Area Under Curve , Blood Glucose/metabolism , Diet , Gastric Inhibitory Polypeptide/metabolism , Glucose Tolerance Test , Glycosuria/metabolism , Methylglucosides/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Postprandial Period/physiology , Sodium-Glucose Transporter 1/genetics , Sodium-Glucose Transporter 1/physiology , Sodium-Glucose Transporter 2/genetics
19.
J Anim Sci ; 91(4): 1707-18, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23345556

ABSTRACT

Probiotics have been shown to have positive effects on growth performance traits and the health of farm animals. The objective of the study was to examine whether the probiotic strain Enterococcus faecium NCIMB 10415 (E. faecium) changes the absorptive and secretory transport and barrier properties of piglet jejunum in vitro and thereby to verify tendencies observed in a former feeding trial with E. faecium. Further aims were to assess a potential mechanism of probiotics by testing effects of IL-α, which is upregulated in the peripheral blood mononuclear cells of E. faecium-supplemented piglets, and to test the hypothesis that IL-1α induces a change in ion transport. Sows and their piglets were randomly assigned to a control group and a probiotic group supplemented with E. faecium. The sows received the probiotic supplemented feed from d 28 before parturition and the piglets from d 12 after birth. Piglets were killed at the age of 12 ± 1, 26 ± 1, 34 ± 1, and 54 ± 1 d. Ussing chamber studies were conducted with isolated mucosae from the mid jejunum. Samples were taken for mRNA expression analysis of sodium-glucose-linked transporter 1 (SGLT1) and cystic fibrosis transmembrane conductance regulator (CFTR). The Na(+)/glucose cotransport was increased in the probiotic group compared with the control group at 26 (P = 0.04) and 54 d of age (P = 0.01). The PGE2-induced short circuit current (Isc) was greater at 54 d of age in the probiotic group compared with the control group (P = 0.03). In addition, effects of age on the absorptive (P < 0.01) and secretory (P < 0.01) capacities were observed. Neither SGLT1 nor CFTR mRNA expression was changed by probiotic supplementation. Mannitol flux rates as a marker of paracellular permeability decreased in both groups with increasing age and were less in the probiotic group at the 26 d of age (P = 0.04), indicating a tighter intestinal barrier. The ΔIsc induced by IL-1α was inhibited by bumetanide (P < 0.01), indicating an induction of Cl(-) secretion. Thus, in this experimental setup, E. faecium increased the absorptive and secretory capacity of jejunal mucosae and enhanced the intestinal barrier. Furthermore, the results indicated that IL-1α induces bumetanide-sensitive chloride secretion. The effects of cytokines as potential mediators of probiotic effects should, therefore, be the subject of further studies.


Subject(s)
Enterococcus faecium , Intestinal Absorption/physiology , Intestinal Mucosa/physiology , Probiotics , Animals , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Electric Impedance , Female , Interleukin-1alpha/physiology , Intestinal Mucosa/microbiology , Jejunum/microbiology , Jejunum/physiology , Male , Mannitol/metabolism , Real-Time Polymerase Chain Reaction/veterinary , Sodium-Glucose Transporter 1/physiology , Swine/microbiology , Swine/physiology
SELECTION OF CITATIONS
SEARCH DETAIL