Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
J Mol Biol ; 434(5): 167464, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35077764

RESUMO

Sodium-glucose cotransporters (SGLTs) are responsible for sugar absorption in small intestine and renal tubule epithelial cells. These proteins have attracted clinical attention as a cause of malabsorption and as a target for diabetes drugs. Each SGLT isoform has strict selectivity for its monosaccharide substrate. Few studies have attempted to elucidate the structural basis of sugar selectivity by allowing generating SGLT mutants that bind substrates not normally transported or by reproducing the substrate specificity of other isoforms. In this study, we built a structural homology model for the substrate binding states of human SGLT1 (hSGLT1), which primarily transports glucose and galactose. We also performed electrophysiological analysis of hSGLT1 using various natural sugars and mutants. By mutating the K321 residue, which forms hydrophilic interactions in the sugar binding pocket, we induced mannose and allose transport. We also changed the glucose/galactose transport ratio, which reproduces the substrate specificity of the prokaryotic galactose transporter. By adding mutations one-by-one to the residues in the binding pocket, we were able to reproduce the substrate specificity of SGLT4, which transports fructose. This suggests that fructose, which exhibits various structures in equilibrium, binds to SGLT in a pyranose conformation. These results reveal one state of the structural basis that determines selective transport by SGLT. These findings will be useful for predicting the substrates of other glucose transporters and to design effective inhibitors.


Assuntos
Transportador 1 de Glucose-Sódio , Transporte Biológico , Frutose/metabolismo , Galactose/metabolismo , Glucose/metabolismo , Humanos , Modelos Moleculares , Mutação , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Transportador 1 de Glucose-Sódio/química , Transportador 1 de Glucose-Sódio/genética , Transportador 1 de Glucose-Sódio/metabolismo , Especificidade por Substrato
2.
Nature ; 601(7892): 274-279, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34880492

RESUMO

Glucose is a primary energy source in living cells. The discovery in 1960s that a sodium gradient powers the active uptake of glucose in the intestine1 heralded the concept of a secondary active transporter that can catalyse the movement of a substrate against an electrochemical gradient by harnessing energy from another coupled substrate. Subsequently, coupled Na+/glucose transport was found to be mediated by sodium-glucose cotransporters2,3 (SGLTs). SGLTs are responsible for active glucose and galactose absorption in the intestine and for glucose reabsorption in the kidney4, and are targeted by multiple drugs to treat diabetes5. Several members within the SGLT family transport key metabolites other than glucose2. Here we report cryo-electron microscopy structures of the prototypic human SGLT1 and a related monocarboxylate transporter SMCT1 from the same family. The structures, together with molecular dynamics simulations and functional studies, define the architecture of SGLTs, uncover the mechanism of substrate binding and selectivity, and shed light on water permeability of SGLT1. These results provide insights into the multifaceted functions of SGLTs.


Assuntos
Microscopia Crioeletrônica , Glucose , Glucose/metabolismo , Humanos , Transportadores de Ácidos Monocarboxílicos/química , Transportadores de Ácidos Monocarboxílicos/metabolismo , Transportadores de Ácidos Monocarboxílicos/ultraestrutura , Sódio/metabolismo , Transportador 1 de Glucose-Sódio/química , Transportador 1 de Glucose-Sódio/metabolismo , Transportador 1 de Glucose-Sódio/ultraestrutura , Especificidade por Substrato
3.
Sci Rep ; 11(1): 13962, 2021 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-34234240

RESUMO

Fish has poor utilization capacity for glucose metabolism. The possible reasons are related to the core regulatory elements of glucose metabolism: transport proteins. Studies on the species and functions of Sglt1 in aquatic animals are scarce, therefore further studies are needed. In this study, the full length of blunt snout bream (Megalobrama amblycephala) sglt1 (Masglt1) was 2965 bp including 5'-UTR region of 168 bp and a 3'-UTR region of 820 bp. Masglt1 have a highest sequence homology in Cypriniformes fish. MaSglt1 protein was identified as a transmembrane protein with 14 α-helix structures locating plasma membrane by the methods of predicted tertiary structure and immunohistochemical staining. MaSglt1 protein has a hollow channel forms which could be specifically coupled with two Na+ ions to recognize glucose and carry out transmembrane transport. High sglt1 mRNA was found in the intestine and kidney. The mRNA levels of intestinal sglt1 had a positive correlation with dietary starch levels at 3 h after feeding, and the mRNA was significantly higher than that at 24 h, however, the mRNA levels of renal sglt1 presented results opposite to those of intestinal sglt1. The mRNA levels of renal sglt1 had a positive correlation with dietary starch levels at 24 h after feeding, and the expression was significantly higher than that at 3 h. These results confirmed that Masglt11 was mainly found in the intestine and kidney and was located in the cell membrane, playing a role in glucose homeostasis.


Assuntos
Fenômenos Fisiológicos da Nutrição Animal , Cipriniformes/genética , Cipriniformes/metabolismo , Transportador 1 de Glucose-Sódio/genética , Transportador 1 de Glucose-Sódio/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , Biologia Computacional/métodos , Cipriniformes/classificação , Expressão Gênica , Imuno-Histoquímica , Modelos Moleculares , Filogenia , Conformação Proteica , Transportador 1 de Glucose-Sódio/química , Relação Estrutura-Atividade
4.
Biomed Res Int ; 2021: 5550180, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33763471

RESUMO

Diabetes mellitus is the most common chronic disorder and leading cause of renal, neurological, and gastrointestinal manifestations in developed and developing countries. Despite of many drugs and combinational therapies, the complications of diabetes are still listed due to severe consequences of those drugs. In past few years, plant-derived drugs draw special attention due to their higher efficacy and fewer side-effects. Momordica charantia also known as bitter melon is referred as an antidiabetic and hypoglycemic plant in native populations of Asia and East Africa. In current study, an in silico approach was used to evaluate the interactions and binding patterns of plant-derived peptides devised from a hypoglycemic protein adMc1 of M. charantia as potential inhibitor of DPP-IV, SGLT1, and GLUT2 receptor proteins. The study has described a novel approach to investigate hypoglycemic peptides to cure diabetes. A total of eighty tetra-, penta-, and hexapeptides were devised from conserved regions of adMc1 homologs. The molecular docking approach using MOE software was employed to reveal inhibiting potentials of devised peptides against three selected proteins. Out of 30 shortlisted ligands six peptides (i.e. SMCG, DECC, TTIT, RTTI, ARNL and TVEV) accomplished the criteria of being good drug candidates against selected receptor proteins following the drugability assessment test. The overall results are acceptable on the basis of ADMET profiling for being good drug candidates against selected proteins.


Assuntos
Dipeptidil Peptidase 4/química , Inibidores da Dipeptidil Peptidase IV/química , Transportador de Glucose Tipo 2 , Hipoglicemiantes/química , Momordica charantia/química , Peptídeos/química , Proteínas de Plantas/química , Transportador 1 de Glucose-Sódio , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Inibidores da Dipeptidil Peptidase IV/uso terapêutico , Transportador de Glucose Tipo 2/antagonistas & inibidores , Transportador de Glucose Tipo 2/química , Humanos , Hipoglicemiantes/uso terapêutico , Peptídeos/uso terapêutico , Transportador 1 de Glucose-Sódio/antagonistas & inibidores , Transportador 1 de Glucose-Sódio/química
5.
Pflugers Arch ; 472(9): 1207-1248, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32829466

RESUMO

Absorption of monosaccharides is mainly mediated by Na+-D-glucose cotransporter SGLT1 and the facititative transporters GLUT2 and GLUT5. SGLT1 and GLUT2 are relevant for absorption of D-glucose and D-galactose while GLUT5 is relevant for D-fructose absorption. SGLT1 and GLUT5 are constantly localized in the brush border membrane (BBM) of enterocytes, whereas GLUT2 is localized in the basolateral membrane (BLM) or the BBM plus BLM at low and high luminal D-glucose concentrations, respectively. At high luminal D-glucose, the abundance SGLT1 in the BBM is increased. Hence, D-glucose absorption at low luminal glucose is mediated via SGLT1 in the BBM and GLUT2 in the BLM whereas high-capacity D-glucose absorption at high luminal glucose is mediated by SGLT1 plus GLUT2 in the BBM and GLUT2 in the BLM. The review describes functions and regulations of SGLT1, GLUT2, and GLUT5 in the small intestine including diurnal variations and carbohydrate-dependent regulations. Also, the roles of SGLT1 and GLUT2 for secretion of enterohormones are discussed. Furthermore, diseases are described that are caused by malfunctions of small intestinal monosaccharide transporters, such as glucose-galactose malabsorption, Fanconi syndrome, and fructose intolerance. Moreover, it is reported how diabetes, small intestinal inflammation, parental nutrition, bariatric surgery, and metformin treatment affect expression of monosaccharide transporters in the small intestine. Finally, food components that decrease D-glucose absorption and drugs in development that inhibit or downregulate SGLT1 in the small intestine are compiled. Models for regulations and combined functions of glucose transporters, and for interplay between D-fructose transport and metabolism, are discussed.


Assuntos
Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Enteropatias/metabolismo , Intestino Delgado/metabolismo , Transportador 1 de Glucose-Sódio/metabolismo , Animais , Proteínas Facilitadoras de Transporte de Glucose/química , Proteínas Facilitadoras de Transporte de Glucose/genética , Humanos , Absorção Intestinal , Transportador 1 de Glucose-Sódio/química , Transportador 1 de Glucose-Sódio/genética
6.
Methods Mol Biol ; 2168: 73-103, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33582988

RESUMO

Here, we present a protocol for the functional characterization of the H+-coupled human peptide transporter PepT1 and sufficient notes to transfer the protocol to the Na+-coupled sugar transporter SGLT1, the organic cation transporter OCT2, the Na+/Ca2+ exchanger NCX, and the neuronal glutamate transporter EAAT3.The assay was developed for the commercially available SURFE2R N1 instrument (Nanion Technologies GmbH) which applies solid supported membrane (SSM)-based electrophysiology. This technique is widely used for the functional characterization of membrane transporters with more than 100 different transporters characterized so far. The technique is cost-effective, easy to use, and capable of high-throughput measurements.SSM-based electrophysiology utilizes SSM-coated gold sensors to physically adsorb membrane vesicles containing the protein of interest. A fast solution exchange provides the substrate and activates transport. For the measurement of PepT1 activity, we applied a peptide concentration jump to activate H+/peptide symport. Proton influx charges the sensor. A capacitive current is measured reflecting the transport activity of PepT1 . Multiple measurements on the same sensor allow for comparison of transport activity under different conditions. Here, we determine EC50 for PepT1-mediated glycylglycine transport and perform an inhibition experiment using the specific peptide inhibitor Lys[Z(NO2)]-Val.


Assuntos
Técnicas Biossensoriais/métodos , Membrana Celular/metabolismo , Eletrofisiologia/métodos , Transportador 1 de Peptídeos/metabolismo , Transportador 1 de Glucose-Sódio/metabolismo , Transporte Biológico , Humanos , Transportador 1 de Peptídeos/química , Transportador 1 de Glucose-Sódio/química
7.
Molecules ; 24(22)2019 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-31698833

RESUMO

Twelve terpenoids were evaluated in the treatment of type 2 diabetes mellitus: seven monoterpenes (geranyl acetate (1), geranic acid (2), citral (3), geraniol (4), methyl geranate (5), nerol (6), and citronellic acid (7)), three sesquiterpenes (farnesal (8), farnesol (9), and farnesyl acetate (10)), one diterpene (geranylgeraniol (11)), and one triterpene (squalene (12)) were selected to carry out a study on normoglycemic and streptozotocin-induced diabetic mice. Among these, 2, 3, 7, 8, 9, and 10 showed antihyperglycemic activity in streptozotocin-induced diabetic mice. They were then selected for evaluation in oral sucrose and lactose tolerance tests (OSTT and OLTT) as well as in an oral glucose tolerance test (OGTT). In the OSTT and OLTT, compounds 3, 7, 8, 9, and 10 showed a reduction in postprandial glucose peaks 2 h after a sucrose or lactose load (comparable to acarbose). In the case of the OGTT, 2, 7, 8, 9, and 10 showed a reduction in postprandial glucose peaks 2 h after a glucose load (comparable to canagliflozin). Our results suggest that the control of postprandial hyperglycemia may be mediated by the inhibition of disaccharide digestion, such as sucrose and lactose, and the regulation of the absorption of glucose. The first case could be associated with an ∝ -glucosidase inhibitory effect and the second with an inhibition of the sodium-glucose type 1 (SGLT-1) cotransporter. Finally, five acyclic terpenes may be candidates for the development and search for new α-glucosidase and SGLT-1 cotransporter inhibitors.


Assuntos
Glicemia , Terpenos/química , Terpenos/farmacologia , Animais , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/tratamento farmacológico , Inibidores de Glicosídeo Hidrolases/química , Inibidores de Glicosídeo Hidrolases/farmacologia , Hipoglicemiantes/química , Hipoglicemiantes/farmacologia , Masculino , Camundongos , Estrutura Molecular , Transportador 1 de Glucose-Sódio/antagonistas & inibidores , Transportador 1 de Glucose-Sódio/química , Relação Estrutura-Atividade , Terpenos/sangue , alfa-Glucosidases/química , alfa-Glucosidases/metabolismo
8.
Pharmacol Res Perspect ; 7(4): e00504, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31384471

RESUMO

Selective analogs of the natural glycoside phloridzin are marketed drugs that reduce hyperglycemia in diabetes by inhibiting the active sodium glucose cotransporter SGLT2 in the kidneys. In addition, intestinal SGLT1 is now recognized as a target for glycemic control. To expand available type 2 diabetes remedies, we aimed to find novel SGLT1 inhibitors beyond the chemical space of glycosides. We screened a bioactive compound library for SGLT1 inhibitors and tested primary hits and additional structurally similar molecules on SGLT1 and SGLT2 (SGLT1/2). Novel SGLT1/2 inhibitors were discovered in separate chemical clusters of natural and synthetic compounds. These have IC50-values in the 10-100 µmol/L range. The most potent identified novel inhibitors from different chemical clusters are (SGLT1-IC50 Mean ± SD, SGLT2-IC50 Mean ± SD): (+)-pteryxin (12 ± 2 µmol/L, 9 ± 4 µmol/L), (+)-ε-viniferin (58 ± 18 µmol/L, 110 µmol/L), quinidine (62 µmol/L, 56 µmol/L), cloperastine (9 ± 3 µmol/L, 9 ± 7 µmol/L), bepridil (10 ± 5 µmol/L, 14 ± 12 µmol/L), trihexyphenidyl (12 ± 1 µmol/L, 20 ± 13 µmol/L) and bupivacaine (23 ± 14 µmol/L, 43 ± 29 µmol/L). The discovered natural inhibitors may be further investigated as new potential (prophylactic) agents for controlling dietary glucose uptake. The new diverse structure activity data can provide a starting point for the optimization of novel SGLT1/2 inhibitors and support the development of virtual SGLT1/2 inhibitor screening models.


Assuntos
Produtos Biológicos/farmacologia , Diabetes Mellitus Tipo 2/tratamento farmacológico , Bibliotecas de Moléculas Pequenas/farmacologia , Transportador 1 de Glucose-Sódio/metabolismo , Transportador 2 de Glucose-Sódio/metabolismo , Animais , Produtos Biológicos/química , Células CHO , Células CACO-2 , Cumarínicos/química , Cumarínicos/farmacologia , Cricetulus , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Concentração Inibidora 50 , Florizina/análogos & derivados , Quinidina/química , Quinidina/farmacologia , Bibliotecas de Moléculas Pequenas/química , Transportador 1 de Glucose-Sódio/química , Transportador 2 de Glucose-Sódio/química
9.
Proc Natl Acad Sci U S A ; 114(46): E9980-E9988, 2017 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-29087341

RESUMO

In the human sodium glucose cotransporter (hSGLT1) cycle, the protein undergoes conformational changes where the sugar-binding site alternatively faces the external and internal surfaces. Functional site-directed fluorometry was used to probe the conformational changes at the sugar-binding site. Residues (Y290, T287, H83, and N78) were mutated to cysteines. The mutants were expressed in Xenopus laevis oocytes and tagged with environmentally sensitive fluorescent rhodamines [e.g., tetramethylrhodamine (TMR)-thiols]. The fluorescence intensity was recorded as the mutants were driven into different conformations using voltage jumps. Sugar binding and transport by the fluorophore-tagged mutants were blocked, but Na+ binding and the voltage-dependent conformational transitions were unaffected. Structural models indicated that external Na+ binding opened a large aqueous vestibule (600 Å3) leading to the sugar-binding site. The fluorescence of TMR covalently linked to Y290C, T287C, and H83C decreased as the mutant proteins were driven from the inward to the outward open Na+-bound conformation. The time courses of fluorescence changes (milliseconds) were close to the SGLT1 capacitive charge movements. The quench in rhodamine fluorescence indicated that the environment of the chromophores became more polar with opening of the external gates as the protein transitioned from the inward to outward facing state. Structural analyses showed an increase in polar side chains and a decrease in hydrophobic side chains lining the vestibule, and this was reflected in solvation of the chromophore. The results demonstrate the opening and closing of external gates in real time, with the accompanying changes of polarity of the sugar vestibule.


Assuntos
Sítios de Ligação/efeitos dos fármacos , Sítios de Ligação/fisiologia , Domínio Catalítico/fisiologia , Fluorometria/métodos , Técnicas de Patch-Clamp/métodos , Transportador 1 de Glucose-Sódio/química , Transportador 1 de Glucose-Sódio/metabolismo , Animais , Sítios de Ligação/genética , Domínio Catalítico/efeitos dos fármacos , Cisteína , Expressão Gênica , Glucose/metabolismo , Íons/metabolismo , Modelos Animais , Modelos Moleculares , Técnicas de Sonda Molecular , Mutação , Oócitos/metabolismo , Polietilenoglicóis/química , Conformação Proteica , Rodaminas/farmacologia , Sódio/metabolismo , Transportador 1 de Glucose-Sódio/genética , Simportadores/metabolismo , Xenopus laevis
10.
Proc Natl Acad Sci U S A ; 113(44): E6887-E6894, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27791155

RESUMO

Membrane transporters, in addition to their major role as specific carriers for ions and small molecules, can also behave as water channels. However, neither the location of the water pathway in the protein nor their functional importance is known. Here, we map the pathway for water and urea through the intestinal sodium/glucose cotransporter SGLT1. Molecular dynamics simulations using the atomic structure of the bacterial transporter vSGLT suggest that water permeates the same path as Na+ and sugar. On a structural model of SGLT1, based on the homology structure of vSGLT, we identified and mutated residues lining the sugar transport pathway to cysteine. The mutants were expressed in Xenopus oocytes, and the unitary water and urea permeabilities were determined before and after modifying the cysteine side chain with reversible methanethiosulfonate reagents. The results demonstrate that water and urea follow the sugar transport pathway through SGLT1. The changes in permeability, increases or decreases, with side-chain modifications depend on the location of the mutation in the region of external or internal gates, or the sugar binding site. These changes in permeability are hypothesized to be due to alterations in steric hindrance to water and urea, and/or changes in protein folding caused by mismatching of side chains in the water pathway. Water permeation through SGLT1 and other transporters bears directly on the structural mechanism for the transport of polar solutes through these proteins. Finally, in vitro experiments on mouse small intestine show that SGLT1 accounts for two-thirds of the passive water flow across the gut.


Assuntos
Proteínas de Membrana Transportadoras/fisiologia , Transportador 1 de Glucose-Sódio/metabolismo , Água/química , Água/metabolismo , Animais , Aquaporinas/metabolismo , Transporte Biológico/fisiologia , Proteínas de Transporte/metabolismo , Glucose/metabolismo , Mucosa Intestinal/metabolismo , Transporte de Íons/fisiologia , Proteínas de Membrana/metabolismo , Camundongos , Modelos Biológicos , Simulação de Dinâmica Molecular , Mutação , Oócitos/metabolismo , Pressão Osmótica , Permeabilidade , Sódio/metabolismo , Transportador 1 de Glucose-Sódio/química , Ureia/metabolismo , Xenopus/metabolismo
11.
Drug Des Devel Ther ; 10: 2929-2938, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27695290

RESUMO

The concept of cardioprotection through preconditioning against ischemia-reperfusion (I/R) injury is well known and established. However, among different proposed mechanisms regarding the concept of ischemic preconditioning, protein kinase C (PKC)-mediated cardioprotection through ischemic preconditioning plays a key role in myocardial I/R injury. Thus, this study was designed to find the relationship between PKC and sodium glucose transporter 1 (SGLT1) in preconditioning-induced cardioprotection, which is ill reported till now. By applying a multifaceted approach, we demonstrated that PKC activates SGLT1, which curbed oxidative stress and apoptosis against I/R injury. PKC activation enhances cardiac glucose uptake through SGLT1 and seems essential in preventing I/R-induced cardiac injury, indicating a possible cross-talk between PKC and SGLT1.


Assuntos
Infarto do Miocárdio/patologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Proteína Quinase C/metabolismo , Transportador 1 de Glucose-Sódio/metabolismo , Ativação Enzimática , Humanos , Infarto do Miocárdio/prevenção & controle , Traumatismo por Reperfusão Miocárdica/metabolismo , Estresse Oxidativo , Proteína Quinase C/química , Transdução de Sinais , Transportador 1 de Glucose-Sódio/química
12.
PLoS One ; 11(5): e0154589, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27137918

RESUMO

Na-coupled cotransporters are proteins that use the trans-membrane electrochemical gradient of Na to activate the transport of a second solute. The sodium-glucose cotransporter 1 (SGLT1) constitutes a well-studied prototype of this transport mechanism but essential molecular characteristics, namely its quaternary structure and the exact arrangement of the C-terminal transmembrane segments, are still debated. After expression in Xenopus oocytes, human SGLT1 molecules (hSGLT1) were labelled on an externally accessible cysteine residue with a thiol-reactive fluorophore (tetramethylrhodamine-C5-maleimide, TMR). Addition of dipicrylamine (DPA, a negatively-charged amphiphatic fluorescence "quencher") to the fluorescently-labelled oocytes is used to quench the fluorescence originating from hSGLT1 in a voltage-dependent manner. Using this arrangement with a cysteine residue introduced at position 624 in the loop between transmembrane segments 12 and 13, the voltage-dependent fluorescence signal clearly indicated that this portion of the 12-13 loop is located on the external side of the membrane. As the 12-13 loop begins on the intracellular side of the membrane, this suggests that the 12-13 loop is re-entrant. Using fluorescence resonance energy transfer (FRET), we observed that different hSGLT1 molecules are within molecular distances from each other suggesting a multimeric complex arrangement. In agreement with this conclusion, a western blot analysis showed that hSGLT1 migrates as either a monomer or a dimer in reducing and non-reducing conditions, respectively. A systematic mutational study of endogenous cysteine residues in hSGLT1 showed that a disulfide bridge is formed between the C355 residues of two neighbouring hSGLT1 molecules. It is concluded that, 1) hSGLT1 is expressed as a disulfide bridged homodimer via C355 and that 2) a portion of the intracellular 12-13 loop is re-entrant and readily accessible from the extracellular milieu.


Assuntos
Dissulfetos/química , Multimerização Proteica , Transportador 1 de Glucose-Sódio/química , Humanos , Espaço Intracelular/metabolismo , Mutação , Estrutura Quaternária de Proteína , Transportador 1 de Glucose-Sódio/genética , Transportador 1 de Glucose-Sódio/metabolismo
13.
J Biol Chem ; 291(18): 9712-20, 2016 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-26945065

RESUMO

The small intestine is void of aquaporins adept at facilitating vectorial water transport, and yet it reabsorbs ∼8 liters of fluid daily. Implications of the sodium glucose cotransporter SGLT1 in either pumping water or passively channeling water contrast with its reported water transporting capacity, which lags behind that of aquaporin-1 by 3 orders of magnitude. Here we overexpressed SGLT1 in MDCK cell monolayers and reconstituted the purified transporter into proteoliposomes. We observed the rate of osmotic proteoliposome deflation by light scattering. Fluorescence correlation spectroscopy served to assess (i) SGLT1 abundance in both vesicles and plasma membranes and (ii) flow-mediated dilution of an aqueous dye adjacent to the cell monolayer. Calculation of the unitary water channel permeability, pf, yielded similar values for cell and proteoliposome experiments. Neither the absence of glucose or Na(+), nor the lack of membrane voltage in vesicles, nor the directionality of water flow grossly altered pf Such weak dependence on protein conformation indicates that a water-impermeable occluded state (glucose and Na(+) in their binding pockets) lasts for only a minor fraction of the transport cycle or, alternatively, that occlusion of the substrate does not render the transporter water-impermeable as was suggested by computational studies of the bacterial homologue vSGLT. Although the similarity between the pf values of SGLT1 and aquaporin-1 makes a transcellular pathway plausible, it renders water pumping physiologically negligible because the passive flux would be orders of magnitude larger.


Assuntos
Permeabilidade da Membrana Celular , Membrana Celular/metabolismo , Transportador 1 de Glucose-Sódio/metabolismo , Água/metabolismo , Animais , Aquaporina 1/química , Aquaporina 1/genética , Aquaporina 1/metabolismo , Transporte Biológico/fisiologia , Membrana Celular/química , Membrana Celular/genética , Cães , Glucose/química , Glucose/metabolismo , Humanos , Células Madin Darby de Rim Canino , Sódio/química , Sódio/metabolismo , Transportador 1 de Glucose-Sódio/química , Transportador 1 de Glucose-Sódio/genética , Água/química
14.
Biochimie ; 115: 187-93, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26086341

RESUMO

The sodium glucose cotransporter SGLT1 expressed mainly in the intestine and kidney has been explored extensively for understanding the mechanism of sugar cotransport and its inhibition by a classical competitive inhibitor, phlorizin (Pz). It has been shown that inhibition of SGLT1 by Pz involves its interaction followed by major conformational changes in the Pz binding domain (PBD) in C-terminal loop 13. However, the mechanism of Pz inhibition and its interaction with other members of SGLT is not known. In this hypothesis, we performed molecular modeling of SGLT1-loop 13 with Pz and carried out primary sequence analyses and secondary structure predictions to determine qualitatively similar PBDs in C-termini of human SGLT2-4, except for vSGLT, which contains an unstructured short C-terminus. The ranking of predictions of Pz interaction strongly agrees with the following ranking of previously reported Pz inhibition: SGLT2>SGLT1>SGLT4>SGLT3>>vSGLT. In addition, the sugar binding residues were found to be quite conserved among all SGLT members investigated here. Based on these preliminary analyses, we propose that other Pz-sensitive SGLTs are also inhibited via mechanism similar to SGLT1 where an aglucone of Pz, phloretin, interacts with PBD and glucoside moiety with sugar binding residues. Our hypothesis sets the stage for future analyses on investigation of Pz interaction with SGLT family and further suggests that Pz modeling may be explored to design novel inhibitors targeting several SGLT members.


Assuntos
Florizina/metabolismo , Transportador 1 de Glucose-Sódio/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Humanos , Dados de Sequência Molecular , Florizina/farmacologia , Ligação Proteica , Estrutura Terciária de Proteína , Transportador 1 de Glucose-Sódio/antagonistas & inibidores , Transportador 1 de Glucose-Sódio/química , Vibrio parahaemolyticus
15.
J Mol Recognit ; 28(8): 467-79, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25753971

RESUMO

Sodium-dependent glucose cotransporters (SGLTs) play an important role in glucose reabsorption in the kidney and have been identified as promising targets to treat diabetes. Because of the side effects like glucose and galactose malabsorption by targeting SGLT1, highly selective SGLT2 inhibitors are more promising in the treatment of diabetes. To understand the mechanism of selectivity, we conducted selectivity-based three-dimensional quantitative structure-activity relationship studies to highlight the structure requirements for highly selective SGLT2 inhibitors. The best comparative molecular field analysis and comparative molecular similarity indices analysis models showed the noncross-validated coefficient (r(2) ) of 0.967 and 0.943, respectively. The predicted correlation coefficients (r(2) pred ) of 0.974 and 0.938 validated the reliability and predictability of these models. Besides, homology models of SGLT2 and SGLT1 were also constructed to investigate the selective mechanism from structure-based perspective. Molecular dynamics simulation and binding free energy calculation were performed on the systems of a potent and selective compound interacting with SGLT2 and SGLT1 to compare the different binding modes. The simulation results showed that the stretch of the methylthio group on Met241 had an essential effect on the different binding modes between SGLT1 and SGLT2, which was consistent with the three-dimensional quantitative structure-activity relationship analysis. Hydrogen bond analysis and binding free energy calculation revealed that SGLT2 binding complex was more stable and favorable than SGLT1 complex, which was highly correlated with the experimental results. Our obtained results give useful information for the investigation of the inhibitors' selectivity between SGLT2 and SGLT1 and will help for further development of highly selective SGLT2 inhibitors.


Assuntos
Transportador 1 de Glucose-Sódio/antagonistas & inibidores , Inibidores do Transportador 2 de Sódio-Glicose , Sódio/metabolismo , Ligantes , Metionina/química , Metionina/metabolismo , Conformação Molecular , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Relação Quantitativa Estrutura-Atividade , Transportador 1 de Glucose-Sódio/química , Transportador 1 de Glucose-Sódio/metabolismo , Transportador 2 de Glucose-Sódio/química , Transportador 2 de Glucose-Sódio/metabolismo , Vibrio parahaemolyticus
16.
J Biol Chem ; 289(31): 21673-83, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24962566

RESUMO

Single molecule force spectroscopy was employed to investigate the dynamics of the sodium glucose co-transporter (SGLT1) upon substrate and inhibitor binding on the single molecule level. CHO cells stably expressing rbSGLT1 were probed by using atomic force microscopy tips carrying either thioglucose, 2'-aminoethyl ß-d-glucopyranoside, or aminophlorizin. Poly(ethylene glycol) (PEG) chains of different length and varying end groups were used as tether. Experiments were performed at 10, 25 and 37 °C to address different conformational states of SGLT1. Unbinding forces between ligands and SGLT1 were recorded at different loading rates by changing the retraction velocity, yielding binding probability, width of energy barrier of the binding pocket, and the kinetic off rate constant of the binding reaction. With increasing temperature, width of energy barrier and average life time increased for the interaction of SGLT1 with thioglucose (coupled via acrylamide to a long PEG) but decreased for aminophlorizin binding. The former indicates that in the membrane-bound SGLT1 the pathway to sugar translocation involves several steps with different temperature sensitivity. The latter suggests that also the aglucon binding sites for transport inhibitors have specific, temperature-sensitive conformations.


Assuntos
Transportador 1 de Glucose-Sódio/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , Microscopia de Força Atômica , Ligação Proteica , Transportador 1 de Glucose-Sódio/antagonistas & inibidores , Transportador 1 de Glucose-Sódio/química
17.
J Gen Physiol ; 140(4): 361-74, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23008432

RESUMO

The Na(+)/glucose cotransporter (SGLT1) is the archetype of membrane proteins that use the electrochemical Na(+) gradient to drive uphill transport of a substrate. The crystal structure recently obtained for vSGLT strongly suggests that SGLT1 adopts the inverted repeat fold of the LeuT structural family for which several crystal structures are now available. What is largely missing is an accurate view of the rates at which SGLT1 transits between its different conformational states. In the present study, we used simulated annealing to analyze a large set of steady-state and pre-steady-state currents measured for human SGLT1 at different membrane potentials, and in the presence of different Na(+) and α-methyl-d-glucose (αMG) concentrations. The simplest kinetic model that could accurately reproduce the time course of the measured currents (down to the 2 ms time range) is a seven-state model (C(1) to C(7)) where the binding of the two Na(+) ions (C(4)→C(5)) is highly cooperative. In the forward direction (Na(+)/glucose influx), the model is characterized by two slow, electroneutral conformational changes (59 and 100 s(-1)) which represent reorientation of the free and of the fully loaded carrier between inside-facing and outside-facing conformations. From the inward-facing (C(1)) to the outward-facing Na-bound configuration (C(5)), 1.3 negative elementary charges are moved outward. Although extracellular glucose binding (C(5)→C(6)) is electroneutral, the next step (C(6)→C(7)) carries 0.7 positive charges inside the cell. Alignment of the seven-state model with a generalized model suggested by the structural data of the LeuT fold family suggests that electrogenic steps are associated with the movement of the so-called thin gates on each side of the substrate binding site. To our knowledge, this is the first model that can quantitatively describe the behavior of SGLT1 down to the 2 ms time domain. The model is highly symmetrical and in good agreement with the structural information obtained from the LeuT structural family.


Assuntos
Transportador 1 de Glucose-Sódio/metabolismo , Animais , Glucose/metabolismo , Humanos , Ativação do Canal Iônico , Cinética , Potenciais da Membrana , Metilglucosídeos/metabolismo , Simulação de Dinâmica Molecular , Proteínas da Membrana Plasmática de Transporte de Neurotransmissores/química , Conformação Proteica , Alinhamento de Sequência , Sódio/metabolismo , Transportador 1 de Glucose-Sódio/química , Xenopus
18.
Cell Biochem Biophys ; 63(2): 151-8, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22383112

RESUMO

Current advances in structural biology provide valuable insights into structure-function relationship of membrane transporters by solving crystal structures of bacterial homologs of human transporters. Therefore, scientists consider bacterial transporters as useful structural models for designing of drugs targeted in human diseases. The functional homology between Vibrio parahaemolyticus Na(+)/galactose transporter (vSGLT) and Na(+)/glucose cotransporter SGLT1 has been well established a decade ago. Now the crystal structure of vSGLT is considered quite valuable in explaining not only the cotransport mechanisms, but it also acts as a representative protein in understanding the protein stability and amino acid interactions within the core structure. We investigated the molecular mechanisms of genetic variations in SGLT1 that cause glucose-galactose malabsorption (GGM) defects using the crystal structure of vSGLT as a model sugar transporter. Our in silico mutagenesis and modeling analysis suggest that the GGM genetic variations lead to conformational changes either by structure destabilization or by formation of unnecessary interaction within the core structure of SGLT1 thereby explaining the genetic defects in Na(+) dependent sugar translocation across the cell membrane.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Galactose/metabolismo , Glucose/metabolismo , Modelos Moleculares , Transportador 1 de Glucose-Sódio/química , Transportador 1 de Glucose-Sódio/genética , Motivos de Aminoácidos , Sequência de Aminoácidos , Substituição de Aminoácidos , Variação Genética , Humanos , Ligação de Hidrogênio , Absorção Intestinal/genética , Síndromes de Malabsorção/genética , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação de Sentido Incorreto , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Homologia Estrutural de Proteína
19.
Biol Trace Elem Res ; 148(2): 224-31, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22354675

RESUMO

Populations chronically exposed to arsenic in drinking water often have increased prevalence of diabetes mellitus. The purpose of this study was to compare the glucose homeostasis of male and female rats exposed to low levels of heavy metals in drinking water. Treated groups were Sprague-Dawley male and female rats exposed to drinking water from Antofagasta city, with total arsenic of 30 ppb and lead of 53 ppb for 3 months; control groups were exposed to purified water by reverse osmosis. The two treated groups in both males and females showed arsenic and lead in the hair of rats. The δ-aminolevulinic acid dehydratase was used as a sensitive biomarker of arsenic toxicity and lead. The activity of δ-aminolevulinic acid dehydratase was reduced only in treated male rats, compared to the control group. Treated males showed a significantly sustained increase in blood glucose and plasma insulin levels during oral glucose tolerance test compared to control group. The oral glucose tolerance test and the homeostasis model assessment of insulin resistance demonstrated that male rats were insulin resistant, and females remained sensitive to insulin after treatment. The total cholesterol and LDL cholesterol increased in treated male rats vs. the control, and triglyceride increased in treated female rats vs. the control. The activity of intestinal Na+/glucose cotransporter in male rats increased compared to female rats, suggesting a significant increase in intestinal glucose absorption. The findings indicate that exposure to low levels of arsenic and lead in drinking water could cause gender differences in insulin resistance.


Assuntos
Arsênio/toxicidade , Água Potável/análise , Exposição Ambiental/análise , Glucose/química , Homeostase , Chumbo/toxicidade , Animais , Intoxicação por Arsênico , Biomarcadores/análise , Glicemia , Chile , LDL-Colesterol/sangue , Água Potável/química , Ativação Enzimática , Eritrócitos/química , Eritrócitos/enzimologia , Feminino , Teste de Tolerância a Glucose , Cabelo/química , Insulina/sangue , Resistência à Insulina , Intestinos/química , Masculino , Sintase do Porfobilinogênio/sangue , Ratos , Ratos Sprague-Dawley , Fatores Sexuais , Transportador 1 de Glucose-Sódio/química , Poluentes Químicos da Água/toxicidade
20.
FEBS Lett ; 586(3): 248-53, 2012 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-22212718

RESUMO

Sodium glucose cotransporters (SGLT) actively catalyse carbohydrate transport across cellular membranes. Six of the 12 known SGLT family members have the capacity to bind and/or transport monosaccharides (SGLT-1 to 6); of these, all but SGLT-5 have been characterised. Here we demonstrate that human SGLT-5 is exclusively expressed in the kidney. Four splice variants were detected and the most abundant SGLT-5-mRNA was functionally characterised. SGLT-5 mediates sodium-dependent [(14)C]-α-methyl-D-glucose (AMG) transport that can be inhibited by mannose, fructose, glucose, and galactose. Uptake studies using demonstrated high capacity transport for mannose and fructose and, to a lesser extent, glucose, AMG, and galactose. SGLT-5 mediated mannose, fructose and AMG transport was weakly (µM potency) inhibited by SGLT-2 inhibitors. In summary, we have characterised SGLT-5 as a kidney mannose transporter. Further studies are warranted to explore the physiological role of SGLT-5.


Assuntos
Rim/metabolismo , Transportador 1 de Glucose-Sódio/metabolismo , Absorção/efeitos dos fármacos , Transporte Biológico/efeitos dos fármacos , Clonagem Molecular , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/metabolismo , Células HEK293 , Humanos , Especificidade de Órgãos , Florizina/farmacologia , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estabilidade Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transportador 1 de Glucose-Sódio/antagonistas & inibidores , Transportador 1 de Glucose-Sódio/química , Transportador 1 de Glucose-Sódio/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...