Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
3.
Diabetologia ; 61(10): 2087-2097, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30132032

RESUMO

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.


Assuntos
Transportador de Glucose Tipo 2/fisiologia , Glucose/metabolismo , Rim/metabolismo , Transportador 1 de Glucose-Sódio/fisiologia , Transportador 2 de Glucose-Sódio/fisiologia , Animais , Glicemia/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Desenho de Fármacos , Transportador de Glucose Tipo 2/genética , Glicosúria/metabolismo , Células HEK293 , Homeostase , Humanos , Hipoglicemiantes/farmacologia , Túbulos Renais/metabolismo , Túbulos Renais Proximais/metabolismo , Camundongos , Camundongos Knockout , Florizina/farmacologia , Transportador 1 de Glucose-Sódio/genética , Transportador 2 de Glucose-Sódio/genética , Inibidores do Transportador 2 de Sódio-Glicose/farmacologia
4.
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
5.
Physiology (Bethesda) ; 32(6): 435-443, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-29021363

RESUMO

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.


Assuntos
Proteínas de Transporte de Sódio-Glucose/genética , Proteínas de Transporte de Sódio-Glucose/metabolismo , Animais , Erros Inatos do Metabolismo dos Carboidratos/genética , Erros Inatos do Metabolismo dos Carboidratos/metabolismo , Glucose/genética , Glucose/metabolismo , Humanos , Síndromes de Malabsorção/genética , Síndromes de Malabsorção/metabolismo
6.
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
7.
Proc Natl Acad Sci U S A ; 113(27): E3960-6, 2016 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-27325773

RESUMO

Secondary active transporters, such as those that adopt the leucine-transporter fold, are found in all domains of life, and they have the unique capability of harnessing the energy stored in ion gradients to accumulate small molecules essential for life as well as expel toxic and harmful compounds. How these proteins couple ion binding and transport to the concomitant flow of substrates is a fundamental structural and biophysical question that is beginning to be answered at the atomistic level with the advent of high-resolution structures of transporters in different structural states. Nonetheless, the dynamic character of the transporters, such as ion/substrate binding order and how binding triggers conformational change, is not revealed from static structures, yet it is critical to understanding their function. Here, we report a series of molecular simulations carried out on the sugar transporter vSGLT that lend insight into how substrate and ions are released from the inward-facing state of the transporter. Our simulations reveal that the order of release is stochastic. Functional experiments were designed to test this prediction on the human homolog, hSGLT1, and we also found that cytoplasmic release is not ordered, but we confirmed that substrate and ion binding from the extracellular space is ordered. Our findings unify conflicting published results concerning cytoplasmic release of ions and substrate and hint at the possibility that other transporters in the superfamily may lack coordination between ions and substrate in the inward-facing state.


Assuntos
Simulação de Dinâmica Molecular , Transportador 1 de Glucose-Sódio/metabolismo , Glucose/metabolismo , Células HEK293 , Humanos , Cadeias de Markov , Método de Monte Carlo , Técnicas de Patch-Clamp , Sódio/metabolismo
8.
Am J Physiol Cell Physiol ; 308(12): C1008-22, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25855082

RESUMO

GLUT12 is a member of the facilitative family of glucose transporters. The goal of this study was to characterize the functional properties of GLUT12, expressed in Xenopus laevis oocytes, using radiotracer and electrophysiological methods. Our results showed that GLUT12 is a facilitative sugar transporter with substrate selectivity: d-glucose ≥ α-methyl-d-glucopyranoside (α-MG) > 2-deoxy-d-glucose(2-DOG) > d-fructose = d-galactose. α-MG is a characteristic substrate of the Na(+)/glucose (SGLT) family and has not been shown to be a substrate of any of the GLUTs. In the absence of sugar, (22)Na(+) was transported through GLUT12 at a higher rate (40%) than noninjected oocytes, indicating that there is a Na(+) leak through GLUT12. Genistein, an inhibitor of GLUT1, also inhibited sugar uptake by GLUT12. Glucose uptake was increased by the PKA activator 8-bromoadenosine 3',5'-cyclic monophosphate (8-Br-cAMP) but not by the PKC activator phorbol-12-myristate-13-acetate (PMA). In high K(+) concentrations, glucose uptake was blocked. Addition of glucose to the external solution induced an inward current with a reversal potential of approximately -15 mV and was blocked by Cl(-) channel blockers, indicating the current was carried by Cl(-) ions. The sugar-activated Cl(-) currents were unaffected by genistein. In high external K(+) concentrations, sugar-activated Cl(-) currents were also blocked, indicating that GLUT12 activity is voltage dependent. Furthermore, glucose-induced current was increased by the PKA activator 8-Br-cAMP but not by the PKC activator PMA. These new features of GLUT12 are very different from those described for other GLUTs, indicating that GLUT12 must have a specific physiological role within glucose homeostasis, still to be discovered.


Assuntos
Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Glucose/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Animais , Transporte Biológico , Canais de Cloreto/antagonistas & inibidores , Canais de Cloreto/metabolismo , Cloretos/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ativação Enzimática , Ativadores de Enzimas/farmacologia , Genisteína/farmacologia , Glucose/análogos & derivados , Proteínas Facilitadoras de Transporte de Glucose/antagonistas & inibidores , Humanos , Concentração de Íons de Hidrogênio , Cinética , Potenciais da Membrana , Oócitos , Técnicas de Patch-Clamp , Sódio/metabolismo , Xenopus laevis
9.
Physiol Rep ; 2(6)2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24973332

RESUMO

SGLT2 inhibitors are a new class of drugs that have been recently developed to treat type II diabetes. They lower glucose levels by inhibiting the renal Na(+)/glucose cotransporter SGLT2, thereby increasing the amount of glucose excreted in the urine. Pharmacodynamics studies have raised questions about how these inhibitors reach SGLT2 in the brush border membrane of the S1 and S2 segments of the renal proximal tubule: are these drugs filtered by the glomerulus and act extracellularly, or do they enter the cell and act intracellularly? To address this question we expressed hSGLT2 in HEK-293T cells and determined the affinity of a specific hSGLT2 inhibitor, TA-3404 (also known as JNJ-30980924), from the extra- and intracellular side of the plasma membrane. Inhibition of SGLT2 activity (Na(+)/glucose currents) by TA-3404 was determined using the whole-cell patch clamp that allows controlling the composition of both the extracellular and intracellular solutions. We compared the results to those obtained using the nonselective SGLT inhibitor phlorizin, and to the effect of TA-3404 on hSGLT1. Our results showed that TA-3404 is a potent extracellular inhibitor of glucose inward SGLT2 transport (IC50 2 nmol/L) but it was ineffective from the intracellular compartment at both low (5 mmol/L) and high (150 mmol/L) intracellular NaCl concentrations. We conclude that TA-3404 only inhibits SGLT2 from the extracellular side of the plasma membrane, suggesting that it is filtered from the blood through the glomerulus and acts from within the tubule lumen.

10.
Am J Physiol Cell Physiol ; 306(9): C864-70, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24573086

RESUMO

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.


Assuntos
Rim/metabolismo , Manose/metabolismo , Proteínas de Transporte de Sódio-Glucose/metabolismo , Sódio/metabolismo , Animais , Cátions , Células HEK293 , Humanos , Concentração de Íons de Hidrogênio , Rim/efeitos dos fármacos , Cinética , Lítio/metabolismo , Potenciais da Membrana , Estrutura Molecular , Florizina/farmacologia , Ratos , Ratos Sprague-Dawley , Proteínas de Transporte de Sódio-Glucose/antagonistas & inibidores , Proteínas de Transporte de Sódio-Glucose/genética , Transfecção
11.
Proc Natl Acad Sci U S A ; 110(47): E4557-66, 2013 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-24191006

RESUMO

Sodium cotransporters from several different gene families belong to the leucine transporter (LeuT) structural family. Although the identification of Na(+) in binding sites is beyond the resolution of the structures, two Na(+) binding sites (Na1 and Na2) have been proposed in LeuT. Na2 is conserved in the LeuT family but Na1 is not. A biophysical method has been used to measure sodium dissociation constants (Kd) of wild-type and mutant human sodium glucose cotransport (hSGLT1) proteins to identify the Na(+) binding sites in hSGLT1. The Na1 site is formed by residues in the sugar binding pocket, and their mutation influences sodium binding to Na1 but not to Na2. For the canonical Na2 site formed by two -OH side chains, S392 and S393, and three backbone carbonyls, mutation of S392 to cysteine increased the sodium Kd by sixfold. This was accompanied by a dramatic reduction in the apparent sugar and phlorizin affinities. We suggest that mutation of S392 in the Na2 site produces a structural rearrangement of the sugar binding pocket to disrupt both the binding of the second Na(+) and the binding of sugar. In contrast, the S393 mutations produce no significant changes in sodium, sugar, and phlorizin affinities. We conclude that the Na2 site is conserved in hSGLT1, the side chain of S392 and the backbone carbonyl of S393 are important in the first Na(+) binding, and that Na(+) binding to Na2 promotes binding to Na1 and also sugar binding.


Assuntos
Sítios de Ligação/genética , Modelos Biológicos , Modelos Moleculares , Conformação Proteica , Transportador 1 de Glucose-Sódio/metabolismo , Sódio/metabolismo , Biofísica/métodos , Humanos , Mutagênese Sítio-Dirigida , Oócitos/metabolismo , Técnicas de Patch-Clamp , Ligação Proteica , Transportador 1 de Glucose-Sódio/genética
12.
Biochemistry ; 51(47): 9480-7, 2012 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-23116249

RESUMO

In the LeuT family of sodium solute symporters, 13-17% of the residues in transmembrane domains are aromatic. The unique properties of aromatic amino acids allow them to play specialized roles in proteins, but their function in membrane transporters is underappreciated. Here we analyze the π bonding pattern in the LeuT (5TMIR) family and then describe the role of a triad of aromatic residues in sodium-dependent sugar cotransporters (SGLTs). In SLC5 symporters, three aromatic residues in TM6 (SGLT1 W289, Y290, and W291) are conserved in only those transporting sugars and inositols. We used biophysical analysis of mutants to discover their functional roles, which we have interpreted in terms of CH-π, π-π, and cation-π bonding. We discovered that (1) glucose binding involves CH-π stacking with Y290, (2) π T-stacking interactions between Y290 and W291 and H-bonding between Y290 and N78 (TM1) are essential to form the sodium and sugar binding sites, (3) the Na(+):sugar stoichiometry is determined by these residues, and (4) W289 may be important in stabilizing the structure through H-bonding to TM3. We also find that the WYW triad plays a role in Na(+) coordination at the Na1 site, possibly through cation-π interactions. Surprisingly, this Na(+) is not necessarily coupled to glucose translocation. Our analysis of π interactions in other LeuT proteins suggests that they also contribute to the structure and function in this whole family of transporters.


Assuntos
Aminoácidos Aromáticos/química , Transportador 1 de Glucose-Sódio/metabolismo , Sódio/metabolismo , Simportadores/química , Motivos de Aminoácidos/fisiologia , Sequência de Aminoácidos , Animais , Sítios de Ligação , Glucose/metabolismo , Humanos , Cinética , Oócitos/fisiologia , Alinhamento de Sequência , Simportadores/genética , Simportadores/metabolismo , Xenopus laevis
13.
Am J Physiol Cell Physiol ; 302(9): C1293-305, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22159082

RESUMO

The Na(+)-glucose cotransporter hSGLT1 is a member of a class of membrane proteins that harness Na(+) electrochemical gradients to drive uphill solute transport. Although hSGLT1 belongs to one gene family (SLC5), recent structural studies of bacterial Na(+) cotransporters have shown that Na(+) transporters in different gene families have the same structural fold. We have constructed homology models of hSGLT1 in two conformations, the inward-facing occluded (based on vSGLT) and the outward open conformations (based on Mhp1), mutated in turn each of the conserved gates and ligand binding residues, expressed the SGLT1 mutants in Xenopus oocytes, and determined the functional consequences using biophysical and biochemical assays. The results establish that mutating the ligand binding residues produces profound changes in the ligand affinity (the half-saturation concentration, K(0.5)); e.g., mutating sugar binding residues increases the glucose K(0.5) by up to three orders of magnitude. Mutation of the external gate residues increases the Na(+) to sugar transport stoichiometry, demonstrating that these residues are critical for efficient cotransport. The changes in phlorizin inhibition constant (K(i)) are proportional to the changes in sugar K(0.5), except in the case of F101C, where phlorizin K(i) increases by orders of magnitude without a change in glucose K(0.5). We conclude that glucose and phlorizin occupy the same binding site and that F101 is involved in binding to the phloretin group of the inhibitor. Substituted-cysteine accessibility methods show that the cysteine residues at the position of the gates and sugar binding site are largely accessible only to external hydrophilic methanethiosulfonate reagents in the presence of external Na(+), demonstrating that the external sugar (and phlorizin) binding vestibule is opened by the presence of external Na(+) and closes after the binding of sugar and phlorizin. Overall, the present results provide a bridge between kinetics and structural studies of cotransporters.


Assuntos
Transportador 1 de Glucose-Sódio/química , Transportador 1 de Glucose-Sódio/metabolismo , Sequência de Aminoácidos , Animais , Humanos , Cinética , Dados de Sequência Molecular , Técnicas de Patch-Clamp , Conformação Proteica , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Relação Estrutura-Atividade , Xenopus laevis
14.
Am J Physiol Cell Physiol ; 302(2): C373-82, 2012 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-21940664

RESUMO

Human Na(+)-D-glucose cotransporter (hSGLT) inhibitors constitute the newest class of diabetes drugs, blocking up to 50% of renal glucose reabsorption in vivo. These drugs have potential for widespread use in the diabetes epidemic, but how they work at a molecular level is poorly understood. Here, we use electrophysiological methods to assess how they block Na(+)-D-glucose cotransporter SGLT1 and SGLT2 expressed in human embryonic kidney 293T (HEK-293T) cells and compared them to the classic SGLT inhibitor phlorizin. Dapagliflozin [(1S)-1,5,5-anhydro-1-C-{4-chloro-3-[(4-ethoxyphenyl)methyl]phenyl}-D-glucitol], two structural analogs, and the aglycones of phlorizin and dapagliflozin were investigated in detail. Dapagliflozin and fluoro-dapagliflozin [(1S)-1,5-anhydro-1-C-{4-chloro-3-[(4-ethoxyphenyl)methyl]phenyl}-4-F-4-deoxy-D-glucitol] blocked glucose transport and glucose-coupled currents with ≈100-fold specificity for hSGLT2 (K(i) = 6 nM) over hSGLT1 (K(i) = 400 nM). As galactose is a poor substrate for SGLT2, it was surprising that galacto-dapagliflozin [(1S)-1,5-anhydro-1-C-{4-chloro-3-[(4-ethoxyphenyl)methyl]phenyl}-D-galactitol] was a selective inhibitor of hSGLT2, but was less potent than dapagliflozin for both transporters (hSGLT2 K(i) = 25 nM, hSGLT1 K(i) = 25,000 nM). Phlorizin and galacto-dapagliflozin rapidly dissociated from SGLT2 [half-time off rate (t(1/2,Off)) ≈ 20-30 s], while dapagliflozin and fluoro-dapagliflozin dissociated from hSGLT2 at a rate 10-fold slower (t(1/2,Off) ≥ 180 s). Phlorizin was unable to exchange with dapagliflozin bound to hSGLT2. In contrast, dapagliflozin, fluoro-dapagliflozin, and galacto-dapagliflozin dissociated quickly from hSGLT1 (t(1/2,Off) = 1-2 s), and phlorizin readily exchanged with dapagliflozin bound to hSGLT1. The aglycones of phlorizin and dapagliflozin were poor inhibitors of both hSGLT2 and hSGLT1 with K(i) values > 100 µM. These results show that inhibitor binding to SGLTs is composed of two synergistic forces: sugar binding to the glucose site, which is not rigid, and so different sugars will change the orientation of the aglycone in the access vestibule; and the binding of the aglycone affects the binding affinity of the entire inhibitor. Therefore, the pharmacophore must include variations in both the structure of the sugar and the aglycone.


Assuntos
Glucosídeos , Florizina , Transportador 1 de Glucose-Sódio/antagonistas & inibidores , Inibidores do Transportador 2 de Sódio-Glicose , Compostos Benzidrílicos , Transporte Biológico/fisiologia , Diabetes Mellitus/metabolismo , Glucose/metabolismo , Glucosídeos/química , Glucosídeos/metabolismo , Células HEK293 , Humanos , Rim/metabolismo , Estrutura Molecular , Técnicas de Patch-Clamp , Florizina/química , Florizina/metabolismo , Transportador 1 de Glucose-Sódio/química , Transportador 1 de Glucose-Sódio/metabolismo , Transportador 2 de Glucose-Sódio/química , Transportador 2 de Glucose-Sódio/metabolismo , Especificidade por Substrato
15.
Physiol Rev ; 91(2): 733-94, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21527736

RESUMO

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.


Assuntos
Proteínas de Transporte de Sódio-Glucose/genética , Proteínas de Transporte de Sódio-Glucose/fisiologia , Sequência de Aminoácidos , Animais , Clonagem Molecular , Regulação da Expressão Gênica , Humanos , Cinética , Modelos Estatísticos , Dados de Sequência Molecular , Estrutura Secundária de Proteína , Proteínas de Transporte de Sódio-Glucose/biossíntese , Relação Estrutura-Atividade , Especificidade por Substrato
16.
Am J Physiol Cell Physiol ; 300(1): C14-21, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20980548

RESUMO

The human Na(+)/D-glucose cotransporter 2 (hSGLT2) is believed to be responsible for the bulk of glucose reabsorption in the kidney proximal convoluted tubule. Since blocking reabsorption increases urinary glucose excretion, hSGLT2 has become a novel drug target for Type 2 diabetes treatment. Glucose transport by hSGLT2 was studied at 37°C in human embryonic kidney 293T cells using whole cell patch-clamp electrophysiology. We compared hSGLT2 with hSGLT1, the transporter in the straight proximal tubule (S3 segment). hSGLT2 transports with surprisingly similar glucose affinity and lower concentrative power than hSGLT1: Na(+)/D-glucose cotransport by hSGLT2 was electrogenic with apparent glucose and Na(+) affinities of 5 and 25 mM, and a Na(+):glucose coupling ratio of 1; hSGLT1 affinities were 2 and 70 mM and coupling ratio of 2. Both proteins showed voltage-dependent steady-state transport; however, unlike hSGLT1, hSGLT2 did not exhibit detectable pre-steady-state currents in response to rapid jumps in membrane voltage. D-Galactose was transported by both proteins, but with very low affinity by hSGLT2 (≥100 vs. 6 mM). ß-D-Glucopyranosides were either substrates or blockers. Phlorizin exhibited higher affinity with hSGLT2 (K(i) 11 vs. 140 nM) and a lower Off-rate (0.03 vs. 0.2 s⁻¹) compared with hSGLT1. These studies indicate that, in the early proximal tubule, hSGLT2 works at 50% capacity and becomes saturated only when glucose is ≥35 mM. Furthermore, results on hSGLT1 suggest it may play a significant role in the reabsorption of filtered glucose in the late proximal tubule. Our electrophysiological study provides groundwork for a molecular understanding of how hSGLT inhibitors affect renal glucose reabsorption.


Assuntos
Glucose/metabolismo , Rim/fisiologia , Transportador 1 de Glucose-Sódio/metabolismo , Transportador 2 de Glucose-Sódio/metabolismo , Potenciais de Ação , Transporte Biológico Ativo , Isótopos de Carbono/metabolismo , Regulação da Expressão Gênica/fisiologia , Células HEK293 , Humanos , Metilglucosídeos/metabolismo , Florizina/farmacologia , Transportador 1 de Glucose-Sódio/genética , Transportador 2 de Glucose-Sódio/genética , Termodinâmica
17.
J Biol Chem ; 284(24): 16226-16235, 2009 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-19363027

RESUMO

The GABA transporter-1 (GAT-1) has three current-generating modes: GABA-coupled current, Li+-induced leak current, and Na+-dependent transient currents. We earlier hypothesized that Li+ is able to substitute for the first Na+ in the transport cycle and thereby induce a distinct conformation in GAT-1 and that the onset of the Li+-induced leak current at membrane potentials more negative than -50 mV was due to a voltage-dependent conformational change of the Li+-bound transporter. In this study, we set out to verify this hypothesis and seek insight into the structural dynamics underlying the leak current, as well as the sodium-dependent transient currents, by applying voltage clamp fluorometry to tetramethylrhodamine 6-maleimide-labeled GAT-1 expressed in Xenopus laevis oocytes. MTSET accessibility studies demonstrated the presence of two distinct conformations of GAT-1 in the presence of Na+ or Li+. The voltage-dependent fluorescence intensity changes obtained in Li+ buffer correlated with the Li+-induced leak currents, i.e. both were highly voltage-dependent and only present at hyperpolarized potentials (<-50 mV). The transient currents correlated directly with the voltage-dependent fluorescence data obtained in sodium buffer and the associated conformational changes were distinct from those associated with the Li+-induced leak current. The inhibitor potency of SKF89976A of the Li+- versus Na+-bound transporter confirmed the cationic dependence of the conformational occupancy. Our observations suggest that the microdomain situated at the external end of transmembrane I is involved in different conformational changes taking place either during the binding and release of sodium or during the initiation of the Li+-induced leak current.


Assuntos
Proteínas da Membrana Plasmática de Transporte de GABA/química , Proteínas da Membrana Plasmática de Transporte de GABA/fisiologia , Cloreto de Sódio/farmacologia , Animais , Soluções Tampão , Cistina/química , Fluorescência , GABAérgicos/farmacologia , Cloreto de Lítio/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Ácidos Nipecóticos/farmacologia , Oócitos/fisiologia , Técnicas de Patch-Clamp , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Ratos , Xenopus laevis
18.
J Membr Biol ; 223(2): 87-106, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18592293

RESUMO

Drugs are transported by cotransporters with widely different turnover rates. We have examined the underlying mechanism using, as a model system, glucose and indican (indoxyl-beta-D-glucopyranoside) transport by human Na+/glucose cotransporter (hSGLT1). Indican is transported by hSGLT1 at 10% of the rate for glucose but with a fivefold higher apparent affinity. We expressed wild-type hSGLT1 and mutant G507C in Xenopus oocytes and used electrical and optical methods to measure the kinetics of glucose (using nonmetabolized glucose analogue alpha-methyl-D-glucopyranoside, alphaMDG) and indican transport, alone and together. Indican behaved as a competitive inhibitor of alphaMDG transport. To examine protein conformations, we recorded SGLT1 capacitive currents (charge movements) and fluorescence changes in response to step jumps in membrane voltage, in the presence and absence of indican and/or alphaMDG. In the absence of sugar, voltage jumps elicited capacitive SGLT currents that decayed to steady state with time constants (tau) of 3-20 ms. These transient currents were abolished in saturating alphaMDG but only slightly reduced (10%) in saturating indican. SGLT1 G507C rhodamine fluorescence intensity increased with depolarizing and decreased with hyperpolarizing voltages. Maximal fluorescence increased approximately 150% in saturating indican but decreased approximately 50% in saturating alphaMDG. Modeling indicated that the rate-limiting step for indican transport is sugar translocation, whereas for alphaMDG it is dissociation of Na+ from the internal binding sites. The inhibitory effects of indican on alphaMDG transport are due to its higher affinity and a 100-fold lower translocation rate. Our results indicate that competition between substrates and drugs should be taken into consideration when targeting transporters as drug delivery systems.


Assuntos
Transporte Biológico Ativo/fisiologia , Glucose/metabolismo , Glucosídeos/metabolismo , Indicã/análogos & derivados , Transportador 1 de Glucose-Sódio/fisiologia , Animais , Fluorescência , Glucosídeos/farmacologia , Humanos , Indicã/metabolismo , Indicã/farmacologia , Cinética , Metilglucosídeos/metabolismo , Metilglucosídeos/farmacologia , Modelos Biológicos , Oócitos , Conformação Proteica , Xenopus laevis
19.
Am J Physiol Renal Physiol ; 294(6): F1422-32, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18367661

RESUMO

The human proton/oligopeptide cotransporters hPEPT1 and hPEPT2 have been targeted to enhance the bioavailability of drugs and prodrugs. Previously, we established the mechanisms of drug transport by hPEPT1. Here, we extend these studies to hPEPT2. Major variants hPEPT2*1 and hPEPT2*2 were expressed in Xenopus oocytes, and each was examined using radiotracer uptake and electrophysiological methods. Glycylsarcosine (Gly-Sar); the beta-lactam antibiotics ampicillin, amoxicillin, cephalexin, and cefadroxil; and the anti-neoplastics delta-aminolevulinic acid (delta-ALA) and bestatin induced inward currents, indicating that they are transported. Variations in transport rate were due to differences in affinity and in turnover rate: for example, cefadroxil was transported with higher apparent affinity but at a lower maximum velocity than Gly-Sar. Transport rates were highest at pH 5 and decreased significantly as the external pH was increased. Our results strongly suggest that the protein does not operate as a cotransporter in tissues where there is little or no pH gradient, such as choroid plexus, lung, or mammary gland. In the absence of substrates, rapid voltage jumps produced hPEPT2 capacitive currents at pH 7. These transients were significantly reduced at pH 5 but recovered on addition of substrates. The seven-state ordered kinetic model previously proposed for hPEPT1 accounts for the steady-state kinetics of neutral drug and dipeptide transport by hPEPT2. The model also explains the capacitive transients, the striking difference in pre-steady-state behavior between hPEPT2 and hPEPT1, and differences in turnover numbers for Gly-Sar and cefadroxil. No functional differences were found between the common variants hPEPT2*1 and hPEPT2*2.


Assuntos
Antibacterianos/farmacocinética , Cefadroxila/farmacocinética , Dipeptídeos/farmacocinética , Oligopeptídeos/metabolismo , Simportadores/metabolismo , Sequência de Aminoácidos , Ácido Aminolevulínico/farmacocinética , Animais , Antineoplásicos/farmacocinética , Feminino , Humanos , Concentração de Íons de Hidrogênio , Cinética , Potenciais da Membrana/fisiologia , Modelos Biológicos , Dados de Sequência Molecular , Oócitos/fisiologia , Transportador 1 de Peptídeos , Polimorfismo Genético , Estrutura Terciária de Proteína , Prótons , Simportadores/química , Simportadores/genética , Trítio , Xenopus laevis
20.
Biochemistry ; 46(46): 13391-406, 2007 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-17960916

RESUMO

The sodium-dependent glucose cotransporter SGLT1 undergoes a series of voltage- and ligand-induced conformational changes that underlie the cotransport mechanism. In this study we describe how the binding of external Na changes the conformation of the sugar-binding domain, exposing residues that are involved in sugar recognition to the external environment. We constructed 15 individual Cys mutants in the four transmembrane helices (TMHs) that form the sugar binding and translocation domain. Each mutant was functionally characterized for transport kinetics and substrate specificity. Identification of interactions between mutated residues and hydroxyls on the pyranose ring was assessed by comparing the affinities of deoxy sugars to those of glucose. We determined conformation-dependent accessibility to the mutated residues by both a traditional substituted cysteine accessibility method (SCAM) and a new fluorescence binding assay. These data were integrated to orient the helices and construct a framework of residues that comprise the external sugar binding site. We present evidence that R499, Q457, and T460 play a direct role in sugar recognition and that five other residues are indirectly involved in transport. Arranging the four TMHs to account for Na-dependent accessibility and potential for sugar interaction allows us to propose a testable model for the SGLT1 sugar binding site.


Assuntos
Glucose/metabolismo , Transportador 1 de Glucose-Sódio/química , Transportador 1 de Glucose-Sódio/metabolismo , Sódio/metabolismo , Animais , Sítios de Ligação , Cisteína/genética , Cisteína/metabolismo , Humanos , Cinética , Modelos Biológicos , Modelos Moleculares , Mutação , Conformação Proteica , Estrutura Terciária de Proteína , Transportador 1 de Glucose-Sódio/genética , Especificidade por Substrato , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...