Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Mol Sci ; 22(11)2021 Jun 03.
Article in English | MEDLINE | ID: mdl-34205045

ABSTRACT

SGLT-2i's exert direct anti-inflammatory and anti-oxidative effects on resting endothelial cells. However, endothelial cells are constantly exposed to mechanical forces such as cyclic stretch. Enhanced stretch increases the production of reactive oxygen species (ROS) and thereby impairs endothelial barrier function. We hypothesized that the SGLT-2i's empagliflozin (EMPA), dapagliflozin (DAPA) and canagliflozin (CANA) exert an anti-oxidative effect and alleviate cyclic stretch-induced endothelial permeability in human coronary artery endothelial cells (HCAECs). HCAECs were pre-incubated with one of the SGLT-2i's (1 µM EMPA, 1 µM DAPA and 3 µM CANA) for 2 h, followed by 10% stretch for 24 h. HCAECs exposed to 5% stretch were considered as control. Involvement of ROS was measured using N-acetyl-l-cysteine (NAC). The sodium-hydrogen exchanger 1 (NHE1) and NADPH oxidases (NOXs) were inhibited by cariporide, or GKT136901, respectively. Cell permeability and ROS were investigated by fluorescence intensity imaging. Cell permeability and ROS production were increased by 10% stretch; EMPA, DAPA and CANA decreased this effect significantly. Cariporide and GKT136901 inhibited stretch-induced ROS production but neither of them further reduced ROS production when combined with EMPA. SGLT-2i's improve the barrier dysfunction of HCAECs under enhanced stretch and this effect might be mediated through scavenging of ROS. Anti-oxidative effect of SGLT-2i's might be partially mediated by inhibition of NHE1 and NOXs.


Subject(s)
Endothelial Cells/drug effects , Inflammation/drug therapy , Oxidative Stress/drug effects , Sodium-Glucose Transport Proteins/antagonists & inhibitors , Sodium-Hydrogen Exchanger 1/antagonists & inhibitors , Benzhydryl Compounds/pharmacology , Canagliflozin/pharmacology , Cell Membrane Permeability/drug effects , Endothelial Cells/metabolism , Glucosides/pharmacology , Guanidines/pharmacology , Humans , Inflammation/genetics , Inflammation/pathology , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/genetics , Oxidative Stress/genetics , Pyrazoles/pharmacology , Pyridones/pharmacology , Reactive Oxygen Species/metabolism , Sodium-Glucose Transport Proteins/genetics , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Sodium-Hydrogen Exchanger 1/genetics , Stress, Mechanical , Sulfones/pharmacology
2.
Pflugers Arch ; 472(9): 1249-1272, 2020 09.
Article in English | MEDLINE | ID: mdl-32394191

ABSTRACT

The fine-tuning of glucose uptake mechanisms is rendered by various glucose transporters with distinct transport characteristics. In the pancreatic islet, facilitative diffusion glucose transporters (GLUTs), and sodium-glucose cotransporters (SGLTs) contribute to glucose uptake and represent important components in the glucose-stimulated hormone release from endocrine cells, therefore playing a crucial role in blood glucose homeostasis. This review summarizes the current knowledge about cell type-specific expression profiles as well as proven and putative functions of distinct GLUT and SGLT family members in the human and rodent pancreatic islet and further discusses their possible involvement in onset and progression of diabetes mellitus. In context of GLUTs, we focus on GLUT2, characterizing the main glucose transporter in insulin-secreting ß-cells in rodents. In addition, we discuss recent data proposing that other GLUT family members, namely GLUT1 and GLUT3, render this task in humans. Finally, we summarize latest information about SGLT1 and SGLT2 as representatives of the SGLT family that have been reported to be expressed predominantly in the α-cell population with a suggested functional role in the regulation of glucagon release.


Subject(s)
Glucose Transport Proteins, Facilitative/metabolism , Islets of Langerhans/metabolism , Sodium-Glucose Transport Proteins/metabolism , Animals , Glucose Transport Proteins, Facilitative/genetics , Humans , Sodium-Glucose Transport Proteins/genetics
3.
Pflugers Arch ; 472(9): 1385-1399, 2020 09.
Article in English | MEDLINE | ID: mdl-32809061

ABSTRACT

Glucose transporters are essential for the heart to sustain its function. Due to its nature as a high energy-consuming organ, the heart needs to catabolize a huge quantity of metabolic substrates. For optimized energy production, the healthy heart constantly switches between various metabolites in accordance with substrate availability and hormonal status. This metabolic flexibility is essential for the maintenance of cardiac function. Glucose is part of the main substrates catabolized by the heart and its use is fine-tuned via complex molecular mechanisms that include the regulation of the glucose transporters GLUTs, mainly GLUT4 and GLUT1. Besides GLUTs, glucose can also be transported by cotransporters of the sodium-glucose cotransporter (SGLT) (SLC5 gene) family, in which SGLT1 and SMIT1 were shown to be expressed in the heart. This SGLT-mediated uptake does not seem to be directly linked to energy production but is rather associated with intracellular signalling triggering important processes such as the production of reactive oxygen species. Glucose transport is markedly affected in cardiac diseases such as cardiac hypertrophy, diabetic cardiomyopathy and heart failure. These alterations are not only fingerprints of these diseases but are involved in their onset and progression. The present review will depict the importance of glucose transport in healthy and diseased heart, as well as proposed therapies targeting glucose transporters.


Subject(s)
Glucose Transport Proteins, Facilitative/metabolism , Heart Diseases/metabolism , Myocardium/metabolism , Sodium-Glucose Transport Proteins/metabolism , Animals , Glucose Transport Proteins, Facilitative/genetics , Heart Diseases/genetics , Humans , Sodium-Glucose Transport Proteins/genetics
4.
Pflugers Arch ; 472(9): 1401-1406, 2020 09.
Article in English | MEDLINE | ID: mdl-32529300

ABSTRACT

Glucose uptake into lymphocytes is accomplished by non-concentrative glucose carriers of the GLUT family (GLUT1, GLUT3, GLUT4, GLUT6) and/or by the Na+-coupled glucose carrier SGLT1. The latter accumulates glucose against glucose gradients and is still effective at very low extracellular glucose concentrations. Signaling involved in SGLT1 expression and activity includes protein kinase A (PKA), protein kinase C (PKC), serum- and glucocorticoid-inducible kinase (SGK1), AMP-activated kinase (AMPK), and Janus kinases (JAK2 and JAK3). Glucose taken up is partially stored as glycogen. In hypoxic environments, such as in tumors as well as infected and inflamed tissues, lymphocytes depend on energy production from glycogen-dependent glycolysis. The lack of SGLT1 may compromise glycogen storage and thus lymphocyte survival and function in hypoxic tissues. Accordingly, in mice, genetic knockout of sglt1 compromised bacterial clearance following Listeria monocytogenes infection leading to an invariably lethal course of the disease. Whether the effect was due to the lack of sglt1 in lymphocytes or in other cell types still remains to be determined. Clearly, additional experimental effort is required to define the role of glucose transport by GLUTs and particularly by SGLT1 for lymphocyte survival and function, as well as orchestration of the host defense against tumors and bacterial infections.


Subject(s)
Glucose Transport Proteins, Facilitative/metabolism , Lymphocytes/metabolism , Sodium-Glucose Transport Proteins/metabolism , Animals , Glucose/metabolism , Glucose Transport Proteins, Facilitative/genetics , Humans , Protein Kinases/metabolism , Signal Transduction , Sodium-Glucose Transport Proteins/genetics
5.
Biol Reprod ; 102(2): 456-474, 2020 02 14.
Article in English | MEDLINE | ID: mdl-31616913

ABSTRACT

Survival and growth of the bovine conceptus (embryo and associated extraembryonic membranes) are dependent on endometrial secretions or histotroph found in the uterine lumen. Previously, serial embryo transfer was used to classify heifers as high fertile (HF), subfertile (SF), or infertile (IF). Here, we investigated specific histotroph components [glucose, prostaglandins (PGs), and lipids] in the uterine lumen of day 17 pregnant and open fertility-classified heifers. Concentrations of glucose in the uterine lumen were increased by pregnancy but did not differ among fertility-classified heifers. Differences in expression of genes encoding glucose transporters and involved with glycolysis and gluconeogenesis were observed between conceptuses collected from HF and SF heifers. In the uterine lumen, PGE2 and PGF2α were increased by pregnancy, and HF heifers had higher concentrations of PGE2, PGF2α, and 6-keto-PFG1α than SF heifers. Differences were found in expression of genes regulating PG signaling, arachidonic acid metabolism, and peroxisome proliferator-activated receptor signaling among conceptuses and endometrium from fertility-classified heifers. Lipidomics was conducted exclusively in samples from HF heifers, and phosphatidylcholine was the main lipid class that increased in the uterine lumen by pregnancy. Expression of several lipid metabolism genes differed between HF and SF conceptuses, and a number of fatty acids were differentially abundant in the uterine lumen of pregnant HF and SF heifers. These results support the ideas that uterine luminal histotroph impacts conceptus survival and programs its development and is a facet of dysregulated conceptus-endometrial interactions that result in loss of the conceptus in SF cattle during the implantation period of pregnancy establishment.


Subject(s)
Glucose/metabolism , Lipids/analysis , Pregnancy, Animal/metabolism , Prostaglandins/metabolism , Uterus/metabolism , Animals , Cattle , Embryo Implantation/physiology , Embryo Transfer/veterinary , Embryonic Development/physiology , Endometrium/metabolism , Female , Gene Expression , Gluconeogenesis/genetics , Glycolysis/genetics , Pregnancy , Sodium-Glucose Transport Proteins/genetics , Sodium-Glucose Transport Proteins/metabolism
6.
Am J Physiol Regul Integr Comp Physiol ; 318(2): R245-R255, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31746628

ABSTRACT

The mucosal-to-serosal flux of 14C 3-O-methyl-d-glucose was compared against the electrogenic transport of d-glucose across ex vivo intestinal segments of Nile tilapia, rainbow trout, and pig in Ussing chambers. The difference in affinities (Km "fingerprints") between pig flux and electrogenic transport of glucose, and the absence of this difference in tilapia and trout, suggest two absorptive pathways in the pig and one in the fish species examined. More specifically, the total mucosal-to-serosal flux revealed a super high-affinity, high-capacity (sHa/Hc) total glucose transport system in tilapia; a super high-affinity, low-capacity (sHa/Lc) total glucose transport system in trout and a low-affinity, low-capacity (La/Lc) total glucose transport system in pig. Comparatively, electrogenic glucose absorption revealed similar Km in both fish species, with a super high-affinity, high capacity (sHa/Hc) system in tilapia; a super high-affinity/super low-capacity (sHa/sLc) system in trout; but a different Km fingerprint in the pig, with a high-affinity, low-capacity (Ha/Lc) system. This was supported by different responses to inhibitors of sodium-dependent glucose transporters (SGLTs) and glucose transporter type 2 (GLUT2) administered on the apical side between species. More specifically, tilapia flux was inhibited by SGLT inhibitors, but not the GLUT2 inhibitor, whereas trout lacked response to inhibitors. In contrast, the pig responded to inhibition by both SGLT and GLUT2 inhibitors with a higher expression of GLUT2. Altogether, it would appear that two pathways are working together in the pig, allowing it to have continued absorption at high glucose concentrations, whereas this is not present in both tilapia and trout.


Subject(s)
3-O-Methylglucose/metabolism , Fish Proteins/metabolism , Glucose Transporter Type 2/metabolism , Intestinal Absorption , Intestinal Mucosa/metabolism , Jejunum/metabolism , Sodium-Glucose Transport Proteins/metabolism , Animals , Cichlids , Female , Glucose Transporter Type 2/genetics , Membrane Potentials , Oncorhynchus mykiss , Sodium-Glucose Transport Proteins/genetics , Species Specificity , Sus scrofa
7.
Fish Physiol Biochem ; 46(3): 1039-1052, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32062828

ABSTRACT

Glucose and fructose play a central role in the metabolism and cellular homeostasis of organisms. Their absorption is co-mediated by two families of glucose transporters, Na+-coupled glucose co-transporters (SGLTs) and facilitative Na+-independent sugar carriers (GLUTs), in the intestine. However, limited information has been available on these transporters in fish. Therefore, we studied glut2, sglt1, and sglt4 genes in grass carp (Ctenopharyngodon idellus). The full-length cDNAs of glut2 was 2308 bp, with an open reading frame (ORF) of 503 amino acids (AAs). The full-length cDNAs of sglt1 was 2890 bp, with an ORF of 658 AAs. Additionally, the full-length cDNAs of sglt4 was 2090 bp, with an ORF encoding 659 AAs. The three deduced AA sequences showed high homology between grass carp and other cyprinid fish species. Based on homology modeling, three-dimensional models of GLUT2, SGLT1, and SGLT4 proteins were created and transmembrane domains were noted. glut2, sglt1, and sglt4 were abundantly expressed in the anterior and mid intestine. In particular, glut2 was markedly expressed in liver (P < 0.05). Additionally, the results indicated that different stocking densities (0.9 or 5.9 kg m-2) did not alter intestinal section-dependent expression patterns of the three transporter genes. However, high stocking density impacted segmental mRNA expression levels. This work demonstrated that mRNA expression of sugar transporter genes in the fish intestine was segment specific, and crowding stress may affect the activity of intestinal sugar transporters. These results provided new insights into the relationship between crowding stress and intestinal sugar transporters in fish.


Subject(s)
Carps/genetics , Fish Proteins/genetics , Glucose Transporter Type 2/genetics , Sodium-Glucose Transport Proteins/genetics , Amino Acid Sequence , Animals , Aquaculture/methods , Base Sequence , Cloning, Molecular , DNA, Complementary/genetics , Fish Proteins/chemistry , Fructose , Glucose , Glucose Transporter Type 2/chemistry , Intestinal Mucosa/metabolism , Kidney/metabolism , Liver/metabolism , Phylogeny , Sodium-Glucose Transport Proteins/chemistry
8.
Am J Physiol Renal Physiol ; 316(3): F473-F480, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30565998

ABSTRACT

Fructose consumption has increased because of widespread use of high-fructose corn syrup by the food industry. Renal proximal tubules are thought to reabsorb fructose. However, fructose reabsorption (Jfructose) by proximal tubules has not yet been directly demonstrated, nor the effects of dietary fructose on Jfructose. This segment expresses Na+- and glucose-linked transporters (SGLTs) 1, 2, 4, and 5 and glucose transporters (GLUTs) 2 and 5. SGLT4 and -5 transport fructose, but SGLT1 and -2 do not. Knocking out SGLT5 increases urinary fructose excretion. We hypothesize that Jfructose in the S2 portion of the proximal tubule is mediated by luminal entry via SGLT4/5 and basolateral exit by GLUT2 and that it is enhanced by a fructose-enriched diet. We measured Jfructose by proximal straight tubules from rats consuming either tap water (Controls) or 20% fructose (FRU). Basal Jfructose in Controls was 14.1 ± 1.5 pmol·mm-1·min-1. SGLT inhibition with phlorizin reduced Jfructose to 4.9 ± 1.4 pmol·mm-1·min-1 ( P < 0.008), whereas removal of Na+ diminished Jfructose by 86 ± 5% ( P < 0.0001). A fructose-enriched diet increased Jfructose from 12.8 ± 2.5 to 19.3 ± 0.5 pmol·mm-1·min-1, a 51% increase ( P < 0.03). Using immunofluorescence, we detected luminal SGLT4 and SGLT5 and basolateral GLUT2; GLUT5 was undetectable. The expression of apical transporters SGLT4 and SGLT5 was higher in FRU than in Controls [137 ± 10% ( P < 0.01) and 38 ± 14% ( P < 0.04), respectively]. GLUT2 was also elevated by 88 ± 27% ( P < 0.02) in FRU. We conclude that Jfructose by proximal tubules occurs primarily via Na+-linked cotransport processes, and a fructose-enriched diet enhances reabsorption. Transport across luminal and basolateral membranes is likely mediated by SGLT4/5 and GLUT2, respectively.


Subject(s)
Carbohydrate Metabolism/physiology , Dietary Carbohydrates/administration & dosage , Fructose/administration & dosage , Glucose Transporter Type 2/metabolism , Kidney Tubules, Proximal/metabolism , Sodium-Glucose Transport Proteins/metabolism , Administration, Oral , Animals , Carbohydrate Metabolism/drug effects , Glucose Transporter Type 2/genetics , Kidney Tubules, Proximal/drug effects , Male , Rats , Rats, Sprague-Dawley , Sodium-Glucose Transport Proteins/genetics
9.
Physiol Rev ; 91(2): 733-94, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21527736

ABSTRACT

There are two classes of glucose transporters involved in glucose homeostasis in the body, the facilitated transporters or uniporters (GLUTs) and the active transporters or symporters (SGLTs). The energy for active glucose transport is provided by the sodium gradient across the cell membrane, the Na(+) glucose cotransport hypothesis first proposed in 1960 by Crane. Since the cloning of SGLT1 in 1987, there have been advances in the genetics, molecular biology, biochemistry, biophysics, and structure of SGLTs. There are 12 members of the human SGLT (SLC5) gene family, including cotransporters for sugars, anions, vitamins, and short-chain fatty acids. Here we give a personal review of these advances. The SGLTs belong to a structural class of membrane proteins from unrelated gene families of antiporters and Na(+) and H(+) symporters. This class shares a common atomic architecture and a common transport mechanism. SGLTs also function as water and urea channels, glucose sensors, and coupled-water and urea transporters. We also discuss the physiology and pathophysiology of SGLTs, e.g., glucose galactose malabsorption and familial renal glycosuria, and briefly report on targeting of SGLTs for new therapies for diabetes.


Subject(s)
Sodium-Glucose Transport Proteins/genetics , Sodium-Glucose Transport Proteins/physiology , Amino Acid Sequence , Animals , Cloning, Molecular , Gene Expression Regulation , Humans , Kinetics , Models, Statistical , Molecular Sequence Data , Protein Structure, Secondary , Sodium-Glucose Transport Proteins/biosynthesis , Structure-Activity Relationship , Substrate Specificity
10.
Physiology (Bethesda) ; 32(6): 435-443, 2017 11.
Article in English | MEDLINE | ID: mdl-29021363

ABSTRACT

It has been 30 years since the intestinal sodium glucose cotransporter SGLT1 was cloned, and, in the intervening years, there have been many advances that have influenced physiology and medicine. Among the first was that SGLT1 is the founding member of the human gene family SLC5, containing 11 diverse transporters and a glucose sensor. Equally surprising was that SGLTs are members of a structural family of cotransporters and exchangers in different gene families. This led to the conclusion that these proteins operate by a mechanism where transport involves the opening and closing of external and internal gates. The mechanism is shared by a wide variety of transporters in different structural families, e.g., the human facilitated glucose transporters (SLC2) in the huge major facilitator superfamily (MFS). Not surprising is the finding that mutations in Sglt genes cause the rare diseases glucose-galactose-malabsorption (GGM) and familial renal glucosuria (FRG). However, it was not envisaged that SGLT inhibitors would be used to treat diabetes mellitus, and these drugs may be able to treat cancer. Finally, in 2017, we have just learned that SGLT1 may be required to resist infection and to avoid recurrent pregnancy loss.


Subject(s)
Sodium-Glucose Transport Proteins/genetics , Sodium-Glucose Transport Proteins/metabolism , Animals , Carbohydrate Metabolism, Inborn Errors/genetics , Carbohydrate Metabolism, Inborn Errors/metabolism , Glucose/genetics , Glucose/metabolism , Humans , Malabsorption Syndromes/genetics , Malabsorption Syndromes/metabolism
11.
Cancer Sci ; 109(12): 3902-3909, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30281872

ABSTRACT

RegulomeDB is a new tool that can predict the regulatory function of genetic variants. We applied RegulomeDB in selecting putative functional variants and evaluated the relationship between these variants and survival outcomes of surgically resected non-small-cell lung cancer. Among the 244 variants studied, 14 were associated with overall survival (P < 0.05) in the discovery cohort and one variant (rs2257609 C>T) was replicated in the validation cohort. In the combined analysis, rs2257609 C>T was significantly associated with worse overall and disease-free survival under a dominant model (P = 2 × 10-5 and P = 0.001, respectively). rs2257609 is located in the SLC5A10 intron, but RegulomeDB predicted that this variant affected DRG2, not SLC5A10 expression. The expression level of SLC5A10 was not different with the rs2257609 genotype. However, DRG2 expression was different according to the rs2257609 genotype (Ptrend   = 0.03) and was significantly higher in tumor than in non-malignant lung tissues (P = 1 × 10-5 ). Luciferase assay also showed higher promoter activity of DRG2 in samples with the rs2257609 T allele (P < 0.0001). rs2257609 C>T affected DRG2 expression and, thus, influenced the prognosis of early-stage non-small-cell lung cancer. This study was approved by the Institutional Review Broad of Kyungpook National University of Hospital (Approval No. KNUMC 2014-04-210-003).


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , GTP-Binding Proteins/genetics , Gene Expression Profiling/methods , Lung Neoplasms/pathology , Polymorphism, Single Nucleotide , Sodium-Glucose Transport Proteins/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Female , Gene Expression Regulation, Neoplastic , Genetic Association Studies , Genetic Variation , Humans , Introns , Lung Neoplasms/genetics , Male , Neoplasm Staging , Prognosis , Promoter Regions, Genetic , Survival Analysis
12.
Mol Phylogenet Evol ; 120: 307-320, 2018 03.
Article in English | MEDLINE | ID: mdl-29233707

ABSTRACT

Metabolic exchange between cnidarians and their symbiotic dinoflagellates is central to maintaining their mutualistic relationship. Sugars are translocated to the host, while ammonium and nitrate are utilized by the dinoflagellates (Symbiodinium spp.). We investigated membrane protein sequences of each partner to identify potential transporter proteins that move sugars into cnidarian cells and nitrogen products into Symbiodinium cells. We examined the facilitated glucose transporters (GLUT), sodium/glucose cotransporters (SGLT), and aquaporin (AQP) channels in the cnidarian host as mechanisms for sugar uptake, and the ammonium and high-affinity nitrate transporters (AMT and NRT2, respectively) in the algal symbiont as mechanisms for nitrogen uptake. Homologous protein sequences were used for phylogenetic analysis and tertiary structure deductions. In cnidarians, we identified putative glucose transporters of the GLUT family and glycerol transporting AQP proteins, as well as sodium monocarboxylate transporters and sodium myo-inositol cotransporters homologous to SGLT proteins. We hypothesize that cnidarians use GLUT proteins as the primary mechanism for glucose uptake, while glycerol moves into cells by passive diffusion. We also identified putative AMT proteins in several Symbiodinium clades and putative NRT2 proteins only in a single clade. We further observed an upregulation of expressed putative AMT proteins in Symbiodinium, which may have emerged as an adaptation to conditions experienced inside the host cell. This study is the first to identify transporter sequences from a diversity of cnidarian species and Symbiodinium clades, which will be useful for future experimental analyses of the host-symbiont proteome and the nutritional exchange of Symbiodinium cells in hospite.


Subject(s)
Cnidaria/classification , Dinoflagellida/classification , Phylogeny , Animals , Anion Transport Proteins/chemistry , Anion Transport Proteins/classification , Anion Transport Proteins/genetics , Aquaporins/chemistry , Aquaporins/classification , Aquaporins/genetics , Cnidaria/metabolism , Computational Biology , Dinoflagellida/metabolism , Nitrate Transporters , Protein Structure, Tertiary , Sodium-Glucose Transport Proteins/chemistry , Sodium-Glucose Transport Proteins/classification , Sodium-Glucose Transport Proteins/genetics , Symbiosis/physiology
13.
J Biomed Sci ; 24(1): 64, 2017 Aug 31.
Article in English | MEDLINE | ID: mdl-28854935

ABSTRACT

Kidneys play an important role in glucose homeostasis. Renal gluconeogenesis prevents hypoglycemia by releasing glucose into the blood stream. Glucose homeostasis is also due, in part, to reabsorption and excretion of hexose in the kidney.Lipid bilayer of plasma membrane is impermeable for glucose, which is hydrophilic and soluble in water. Therefore, transport of glucose across the plasma membrane depends on carrier proteins expressed in the plasma membrane. In humans, there are three families of glucose transporters: GLUT proteins, sodium-dependent glucose transporters (SGLTs) and SWEET. In kidney, only GLUTs and SGLTs protein are expressed. Mutations within genes that code these proteins lead to different renal disorders and diseases. However, diseases, not only renal, such as diabetes, may damage expression and function of renal glucose transporters.


Subject(s)
Glucose Transport Proteins, Facilitative/genetics , Kidney Diseases/genetics , Kidney/physiology , Sodium-Glucose Transport Proteins/genetics , Glucose Transport Proteins, Facilitative/metabolism , Humans , Sodium-Glucose Transport Proteins/metabolism
14.
J Physiol ; 594(15): 4425-38, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27018980

ABSTRACT

KEY POINTS: Glucose transporters are central players in glucose homeostasis. There are two major classes of glucose transporters in the body, the passive facilitative glucose transporters (GLUTs) and the secondary active sodium-coupled glucose transporters (SGLTs). In the present study, we report the use of a non-invasive imaging technique, positron emission tomography, in mice aiming to evaluate the role of GLUTs and SGLTs in controlling glucose distribution and utilization. We show that GLUTs are most significant for glucose uptake into the brain and liver, whereas SGLTs are important in glucose recovery in the kidney. This work provides further support for the use of SGLT imaging in the investigation of the role of SGLT transporters in human physiology and diseases such as diabetes and cancer. ABSTRACT: The importance of sodium-coupled glucose transporters (SGLTs) and facilitative glucose transporters (GLUTs) in glucose homeostasis was studied in mice using fluorine-18 labelled glucose molecular imaging probes and non-invasive positron emission tomography (PET) imaging. The probes were: α-methyl-4-[F-18]-fluoro-4-deoxy-d-glucopyranoside (Me-4FDG), a substrate for SGLTs; 4-deoxy-4-[F-18]-fluoro-d-glucose (4-FDG), a substrate for SGLTs and GLUTs; and 2-deoxy-2-[F-18]-fluoro-d-glucose (2-FDG), a substrate for GLUTs. These radiolabelled imaging probes were injected i.v. into wild-type, Sglt1(-/-) , Sglt2(-/-) and Glut2(-/-) mice and their dynamic whole-body distribution was determined using microPET. The distribution of 2-FDG was similar to that reported earlier (i.e. it accumulated in the brain, heart, liver and kidney, and was excreted into the urinary bladder). There was little change in the distribution of 2-FDG in Glut2(-/-) mice, apart from a reduction in the rate of uptake into liver. The major differences between Me-4FDG and 2-FDG were that Me-4FDG did not enter the brain and was not excreted into the urinary bladder. There was urinary excretion of Me-4FDG in Sglt1(-/-) and Sglt2(-/-) mice. However, Me-4FDG was not reabsorbed in the kidney in Glut2(-/-) mice. There were no differences in Me-4FDG uptake into the heart of wild-type, Sglt1(-/-) and Sglt2(-/-) mice. We conclude that GLUT2 is important in glucose liver transport and reabsorption of glucose in the kidney along with SGLT2 and SGLT1. Complete reabsorption of Me-4FDG from the glomerular filtrate in wild-type mice and the absence of reabsorption in the kidney in Glut2(-/-) mice confirm the importance of GLUT2 in glucose absorption across the proximal tubule.


Subject(s)
Glucose Transport Proteins, Facilitative/metabolism , Sodium-Glucose Transport Proteins/metabolism , Animals , Brain/diagnostic imaging , Brain/metabolism , Deoxyglucose/analogs & derivatives , Female , Fluorine Radioisotopes , Glucose/pharmacokinetics , Glucose Transport Proteins, Facilitative/genetics , Glucosides , Heart/diagnostic imaging , Kidney/diagnostic imaging , Kidney/metabolism , Liver/diagnostic imaging , Liver/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Muscles/diagnostic imaging , Muscles/metabolism , Myocardium/metabolism , Positron-Emission Tomography , Sodium-Glucose Transport Proteins/genetics , Urinary Bladder/diagnostic imaging , Urinary Bladder/metabolism
15.
Biochim Biophys Acta ; 1838(1 Pt B): 244-53, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23988430

ABSTRACT

The sodium/iodide symporter (NIS or SLC5A5) is an intrinsic membrane protein implicated in iodide uptake into thyroid follicular cells. It plays a crucial role in iodine metabolism and thyroid regulation and its function is widely exploited in the diagnosis and treatment of benign and malignant thyroid diseases. A great effort is currently being made to develop a NIS-based gene therapy also allowing the radiotreatment of nonthyroidal tumors. NIS is also expressed in other tissues, such as salivary gland, stomach and mammary gland during lactation, where its physiological role remains unclear. The molecular identity of the thyroid iodide transporter was elucidated approximately fifteen years ago. It belongs to the superfamily of sodium/solute symporters, SSS (and to the human transporter family, SLC5), and is composed of 13 transmembrane helices and 643 amino acid residues in humans. Knowledge concerning NIS structure/function relationship has been obtained by taking advantage of the high resolution structure of one member of the SSS family, the Vibrio parahaemolyticus sodium/galactose symporter (vSGLT), and from studies of gene mutations leading to congenital iodine transport defects (ITD). This review will summarize current knowledge regarding the molecular characterization of NIS.


Subject(s)
Bacterial Proteins/chemistry , Iodides/chemistry , Sodium-Glucose Transport Proteins/chemistry , Sodium/chemistry , Symporters/chemistry , Thyroid Gland/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Humans , Iodides/metabolism , Ion Transport , Molecular Sequence Data , Protein Structure, Secondary , Protein Structure, Tertiary , Sodium/metabolism , Sodium-Glucose Transport Proteins/genetics , Sodium-Glucose Transport Proteins/metabolism , Structural Homology, Protein , Symporters/genetics , Symporters/metabolism , Thyroid Gland/metabolism , Vibrio parahaemolyticus/chemistry , Vibrio parahaemolyticus/genetics , Vibrio parahaemolyticus/metabolism
16.
Am J Physiol Cell Physiol ; 306(9): C864-70, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24573086

ABSTRACT

Sodium glucose cotransporters (SGLTs) mediate the translocation of carbohydrates across the brush border membrane of different organs such as intestine, kidney, and brain. The human SGLT5 (hSGLT5), in particular, is localized in the kidney were it is responsible for mannose and fructose reabsorption from the glomerular filtrate as confirmed by more recent studies on hSGLT5 knockout mice. Here we characterize the functional properties of hSGLT5 expressed in a stable T-Rex-HEK-293 cell line using biochemical and electrophysiological assays. We confirmed that hSGLT5 is a sodium/mannose transporter that is blocked by phlorizin. Li(+) and H(+) ions were also able to drive mannose transport, and transport was electrogenic. Our results moreover indicate that substrates require a pyranose ring with an axial hydroxyl group (-OH) on carbon 2 (C-2). Compared with Na(+)/glucose cotransport, the level of function of Na(+)/mannose cotransport in rat kidney slices was low.


Subject(s)
Kidney/metabolism , Mannose/metabolism , Sodium-Glucose Transport Proteins/metabolism , Sodium/metabolism , Animals , Cations , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Kidney/drug effects , Kinetics , Lithium/metabolism , Membrane Potentials , Molecular Structure , Phlorhizin/pharmacology , Rats , Rats, Sprague-Dawley , Sodium-Glucose Transport Proteins/antagonists & inhibitors , Sodium-Glucose Transport Proteins/genetics , Transfection
17.
Nutr Metab Cardiovasc Dis ; 24(6): 623-31, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24462364

ABSTRACT

BACKGROUND AND AIMS: Sustained hyperglycemia as a result of diabetes mellitus results in over-expression of glucose transporters (GLUTs/SGLTs), protein kinase C-α (PKC-α) and transforming growth factor-ß (TGF-ß) in kidney which increases synthesis and accumulation of extracellular matrix (ECM) components leading to diabetic nephropathy. Previous results from our laboratory showed that banana flower (BF) and pseudostem (BS) ameliorated diabetic complications and reduced formation of advanced glycation end-products (AGEs). In this study, attempts were made to delineate the changes observed in GLUTs and ECM components in kidney by feeding BF and BS at the molecular level. METHODS AND RESULTS: Diabetes was induced in male Wistar rats by injecting streptozotocin. Rats were fed with standard AIN-76 diet or diet supplemented with 5% BF or BS. Rats fed with diet supplemented with aminoguanidine (0.05%) were used as a positive control. Effect of BF and BS on expression of GLUTs/SGLTs, PKC and TGF ß in kidney was evaluated by RT-PCR and accumulation of ECM components in kidney was quantitated by ELISA and immunohistochemistry. BF and BS modulated the over-expression of GLUT 1, 2, 5, SGLT 1, 2 and factors such as PKC-α and TGF-ß to various extents. This impinged on the synthesis of ECM components like laminin, fibronectin and type-IV collagen. CONCLUSION: The results suggest that BF and BS reduce the diabetic nephropathy complications which are accompanied by changes at the molecular level.


Subject(s)
Diabetes Mellitus, Experimental/diet therapy , Diabetic Nephropathies/prevention & control , Dietary Supplements , Gene Expression Regulation , Hypoglycemic Agents/therapeutic use , Kidney/metabolism , Musa , Animals , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Down-Regulation , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Flowers , Glucose Transport Proteins, Facilitative/genetics , Glucose Transport Proteins, Facilitative/metabolism , Hypertrophy , India , Kidney/enzymology , Kidney/pathology , Male , Plant Stems , Protein Kinase C-alpha/genetics , Protein Kinase C-alpha/metabolism , Rats, Wistar , Sodium-Glucose Transport Proteins/genetics , Sodium-Glucose Transport Proteins/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
18.
Nat Commun ; 15(1): 2359, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38504097

ABSTRACT

Genetic mechanisms of blood pressure (BP) regulation remain poorly defined. Using kidney-specific epigenomic annotations and 3D genome information we generated and validated gene expression prediction models for the purpose of transcriptome-wide association studies in 700 human kidneys. We identified 889 kidney genes associated with BP of which 399 were prioritised as contributors to BP regulation. Imputation of kidney proteome and microRNAome uncovered 97 renal proteins and 11 miRNAs associated with BP. Integration with plasma proteomics and metabolomics illuminated circulating levels of myo-inositol, 4-guanidinobutanoate and angiotensinogen as downstream effectors of several kidney BP genes (SLC5A11, AGMAT, AGT, respectively). We showed that genetically determined reduction in renal expression may mimic the effects of rare loss-of-function variants on kidney mRNA/protein and lead to an increase in BP (e.g., ENPEP). We demonstrated a strong correlation (r = 0.81) in expression of protein-coding genes between cells harvested from urine and the kidney highlighting a diagnostic potential of urinary cell transcriptomics. We uncovered adenylyl cyclase activators as a repurposing opportunity for hypertension and illustrated examples of BP-elevating effects of anticancer drugs (e.g. tubulin polymerisation inhibitors). Collectively, our studies provide new biological insights into genetic regulation of BP with potential to drive clinical translation in hypertension.


Subject(s)
Hypertension , Proteome , Humans , Blood Pressure/genetics , Proteome/genetics , Proteome/metabolism , Transcriptome/genetics , Multiomics , Hypertension/metabolism , Kidney/metabolism , Sodium-Glucose Transport Proteins/genetics , Sodium-Glucose Transport Proteins/metabolism
19.
Hypertension ; 81(6): 1296-1307, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38545789

ABSTRACT

BACKGROUND: A fructose high-salt (FHS) diet increases systolic blood pressure and Ang II (angiotensin II)-stimulated proximal tubule (PT) superoxide (O2-) production. These increases are prevented by scavenging O2- or an Ang II type 1 receptor antagonist. SGLT4 (sodium glucose-linked cotransporters 4) and SGLT5 are implicated in PT fructose reabsorption, but their roles in fructose-induced hypertension are unclear. We hypothesized that PT fructose reabsorption by SGLT5 initiates a genetic program enhancing Ang II-stimulated oxidative stress in males and females, thereby causing fructose-induced salt-sensitive hypertension. METHODS: We measured systolic blood pressure in male and female Sprague-Dawley (wild type [WT]), SGLT4 knockout (-/-), and SGLT5-/- rats. Then, we measured basal and Ang II-stimulated (37 nmol/L) O2- production by PTs and conducted gene coexpression network analysis. RESULTS: In male WT and female WT rats, FHS increased systolic blood pressure by 15±3 (n=7; P<0.0027) and 17±4 mm Hg (n=9; P<0.0037), respectively. Male and female SGLT4-/- had similar increases. Systolic blood pressure was unchanged by FHS in male and female SGLT5-/-. In male WT and female WT fed FHS, Ang II stimulated O2- production by 14±5 (n=6; P<0.0493) and 8±3 relative light units/µg protein/s (n=7; P<0.0218), respectively. The responses of SGTL4-/- were similar. Ang II did not stimulate O2- production in tubules from SGLT5-/-. Five gene coexpression modules were correlated with FHS. These correlations were completely blunted in SGLT5-/- and partially blunted by chronically scavenging O2- with tempol. CONCLUSIONS: SGLT5-mediated PT fructose reabsorption is required for FHS to augment Ang II-stimulated proximal nephron O2- production, and increases in PT oxidative stress likely contribute to FHS-induced hypertension.


Subject(s)
Blood Pressure , Fructose , Hypertension , Kidney Tubules, Proximal , Oxidative Stress , Rats, Sprague-Dawley , Animals , Fructose/pharmacology , Oxidative Stress/drug effects , Male , Female , Rats , Hypertension/metabolism , Hypertension/genetics , Hypertension/chemically induced , Hypertension/physiopathology , Blood Pressure/drug effects , Blood Pressure/physiology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/drug effects , Sodium-Glucose Transport Proteins/genetics , Sodium-Glucose Transport Proteins/metabolism , Sodium Chloride, Dietary/adverse effects , Angiotensin II , Disease Models, Animal
20.
Biochim Biophys Acta ; 1818(2): 263-71, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21978597

ABSTRACT

Employing molecular dynamics (MD) simulations, the pathway and mechanism of substrate unbinding from the inward-facing state of the Na(+)-coupled galactose transporter, vSGLT, have been investigated. During a 200-ns equilibrium simulation, repeated spontaneous unbinding events of the substrate from its binding site have been observed. In contrast to the previously proposed gating role of a tyrosine residue (Y263), the unbinding mechanism captured in the present equilibrium simulation does not rely on the displacement and/or rotation of this side chain. Rather, the unbinding involves an initial lateral displacement of the substrate out of the binding site which allows the substrate to completely emerge from the region covered by the side chain of Y263 without any noticeable conformational changes of the latter. Starting with the snapshots taken from this equilibrium simulation with the substrate outside the binding site, steered MD (SMD) simulations were then used to probe the translocation of the substrate along the remaining of the release pathway within the protein's lumen and to characterize the nature of protein-substrate interactions involved in the process. Combining the results of the equilibrium and SMD simulations, we provide a description of the full translocation pathway for the substrate release from the binding site into the cytoplasm. Residues E68, N142, T431, and N267 facilitate the initial substrate's displacement out of the binding site, while the translocation of the substrate along the remainder of the exit pathway formed between TM6 and TM8 is facilitated by H-bond interactions between the substrate and a series of conserved, polar residues (Y138, N267, R273, S365, S368, N371, S372, and T375). The observed molecular events indicate that no gating is required for the release of the substrate from the crystallographically captured structure of the inward-facing state of SGLT, suggesting that this conformation might represent an open, rather than occluded, state of the transporter. This article is part of a Special Issue entitled: Membrane protein structure and function.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Galactose/metabolism , Sodium-Glucose Transport Proteins/chemistry , Sodium-Glucose Transport Proteins/metabolism , Sodium/metabolism , Bacteria/chemistry , Bacteria/genetics , Bacteria/metabolism , Bacterial Proteins/genetics , Binding Sites , Biological Transport , Molecular Dynamics Simulation , Protein Binding , Sodium-Glucose Transport Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL