Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Am Soc Nephrol ; 28(1): 85-93, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27288013

RESUMO

The renal proximal tubule reabsorbs 90% of the filtered glucose load through the Na+-coupled glucose transporter SGLT2, and specific inhibitors of SGLT2 are now available to patients with diabetes to increase urinary glucose excretion. Using expression cloning, we identified an accessory protein, 17 kDa membrane-associated protein (MAP17), that increased SGLT2 activity in RNA-injected Xenopus oocytes by two orders of magnitude. Significant stimulation of SGLT2 activity also occurred in opossum kidney cells cotransfected with SGLT2 and MAP17. Notably, transfection with MAP17 did not change the quantity of SGLT2 protein at the cell surface in either cell type. To confirm the physiologic relevance of the MAP17-SGLT2 interaction, we studied a cohort of 60 individuals with familial renal glucosuria. One patient without any identifiable mutation in the SGLT2 coding gene (SLC5A2) displayed homozygosity for a splicing mutation (c.176+1G>A) in the MAP17 coding gene (PDZK1IP1). In the proximal tubule and in other tissues, MAP17 is known to interact with PDZK1, a scaffolding protein linked to other transporters, including Na+/H+ exchanger 3, and to signaling pathways, such as the A-kinase anchor protein 2/protein kinase A pathway. Thus, these results provide the basis for a more thorough characterization of SGLT2 which would include the possible effects of its inhibition on colocalized renal transporters.


Assuntos
Rim/metabolismo , Proteínas de Membrana/fisiologia , Transportador 2 de Glucose-Sódio/fisiologia , Animais , Células Cultivadas , Glicosúria Renal/genética , Humanos , Rim/citologia , Túbulos Renais Proximais , Proteínas de Membrana/genética , Mutação , Gambás
2.
Am J Physiol Renal Physiol ; 313(2): F467-F474, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28592437

RESUMO

The cotransporter SGLT2 is responsible for 90% of renal glucose reabsorption, and we recently showed that MAP17 appears to work as a required ß-subunit. We report in the present study a detailed functional characterization of human SGLT2 in coexpression with human MAP17 in Xenopus laevis oocytes. Addition of external glucose generates a large inward current in the presence of Na, confirming an electrogenic transport mechanism. At a membrane potential of -50 mV, SGLT2 affinity constants for glucose and Na are 3.4 ± 0.4 and 18 ± 6 mM, respectively. The change in the reversal potential of the cotransport current as a function of external glucose concentration clearly confirms a 1:1 Na-to-glucose transport stoichiometry. SGLT2 is selective for glucose and α-methylglucose but also transports, to a lesser extent, galactose and 3-O-methylglucose. SGLT2 can be inhibited in a competitive manner by phlorizin (Ki = 31 ± 4 nM) and by dapagliflozin (Ki = 0.75 ± 0.3 nM). Similarly to SGLT1, SGLT2 can be activated by Na, Li, and protons. Pre-steady-state currents for SGLT2 do exist but are small in amplitude and relatively fast (a time constant of ~2 ms). The leak current defined as the phlorizin-sensitive current in the absence of substrate was extremely small in the case of SGLT2. In summary, in comparison with SGLT1, SGLT2 has a lower affinity for glucose, a transport stoichiometry of 1:1, very small pre-steady-state and leak currents, a 10-fold higher affinity for phlorizin, and an affinity for dapagliflozin in the subnanomolar range.


Assuntos
Glucose/metabolismo , Rim/metabolismo , Proteínas de Membrana/metabolismo , Reabsorção Renal , Transportador 2 de Glucose-Sódio/metabolismo , Sódio/metabolismo , 3-O-Metilglucose/metabolismo , Animais , Compostos Benzidrílicos/farmacologia , Transporte Biológico , Relação Dose-Resposta a Droga , Galactose , Glucosídeos/farmacologia , Humanos , Rim/efeitos dos fármacos , Cinética , Potenciais da Membrana , Proteínas de Membrana/genética , Metilglucosídeos/metabolismo , Florizina/farmacologia , Reabsorção Renal/efeitos dos fármacos , Transportador 2 de Glucose-Sódio/genética , Inibidores do Transportador 2 de Sódio-Glicose , Xenopus laevis
3.
N Engl J Med ; 362(12): 1102-9, 2010 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-20335586

RESUMO

We describe two siblings from a consanguineous family with autosomal recessive Fanconi's syndrome and hypophosphatemic rickets. Genetic analysis revealed a homozygous in-frame duplication of 21 bp in SLC34A1, which encodes the renal sodium-inorganic phosphate cotransporter NaPi-IIa, as the causative mutation. Functional studies in Xenopus laevis oocytes and in opossum kidney cells indicated complete loss of function of the mutant NaPi-IIa, resulting from failure of the transporter to reach the plasma membrane. These findings show that disruption of the human NaPi-IIa profoundly impairs overall renal phosphate reabsorption and proximal-tubule function and provide evidence of the critical role of NaPi-IIa in human renal phosphate handling.


Assuntos
Raquitismo Hipofosfatêmico Familiar/genética , Síndrome de Fanconi/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/genética , Adulto , Animais , Calcitriol/sangue , Células Cultivadas , Consanguinidade , Análise Mutacional de DNA , Feminino , Genes Recessivos , Humanos , Rim/citologia , Rim/metabolismo , Masculino , Mutação , Oócitos/metabolismo , Gambás , Linhagem , Irmãos , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/metabolismo , Xenopus laevis
4.
Biophys J ; 98(2): 231-9, 2010 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-20338844

RESUMO

Expression of the Na(+)/glucose cotransporter SGLT1 in Xenopus oocytes is characterized by a phlorizin-sensitive leak current (in the absence of glucose) that was originally called a "Na(+) leak" and represents some 5-10% of the maximal Na(+)/glucose cotransport current. We analyzed the ionic nature of the leak current using a human SGLT1 mutant (C292A) displaying a threefold larger leak current while keeping a reversal potential (V(R)) of approximately -15 mV as observed for wt SGLT1. V(R) showed only a modest negative shift when extracellular Na(+) concentration ([Na(+)](o)) was lowered and it was completely insensitive to changes in extracellular Cl(-). When extracellular pH (pH(o)) was decreased from 7.5 to 6.5 and 5.5, V(R) shifted by +15 and +40 mV, respectively, indicating that protons may be the main charge carrier at low pH(o) but other ions must be involved at pH(o) 7.5. In the presence of 15 mM [Na(+)](o) (pH(o) = 7.5), addition of 75 mM of either Na(+), Li(+), Cs(+), or K(+) generated similar increases in the leak current amplitude. This observation, which was confirmed with wt SGLT1, indicates a separate pathway for the leak current with respect to the cotransport current. This means that, contrary to previous beliefs, the leak current cannot be accounted for by the translocation of the Na-loaded and glucose-free cotransporter. Using chemical modification and different SGLT1 mutants, a relationship was found between the cationic leak current and the passive water permeability suggesting that water and cations may share a common pathway through the cotransporter.


Assuntos
Íons/química , Transportador 1 de Glucose-Sódio/química , Animais , Césio/química , Cloretos/química , Ditiotreitol/química , Espaço Extracelular/química , Humanos , Concentração de Íons de Hidrogênio , Lítio/química , Potenciais da Membrana , Mutação de Sentido Incorreto , Técnicas de Patch-Clamp , Potássio/química , Substâncias Redutoras/química , Sódio/química , Transportador 1 de Glucose-Sódio/genética , Água/química , Xenopus laevis
5.
Am J Physiol Cell Physiol ; 298(1): C124-31, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19864324

RESUMO

SMCT1 is a Na-coupled cotransporter of short chain monocarboxylates, which is expressed in the apical membrane of diverse epithelia such as colon, renal cortex, and thyroid. We previously reported that SMCT1 cotransport was reduced by extracellular Cl(-) replacement with cyclamate(-) and that the protein exhibited an ostensible anionic leak current. In this paper, we have revisited the interaction between small monovalent anions and SMCT cotransport and leak currents. We found that the apparent Cl(-) dependence of cotransport was due to inhibition of this protein by the replacement anion cyclamate, whereas several other replacement anions function as substrates for SMCT1; a suitable replacement anion (MES(-)) was identified. The observed outward leak currents represented anionic influx and favored larger anions (NO(3)(-)>I(-)>Br(-)>Cl(-)); currents in excess of 1 muA (at +50 mV) could be observed and exhibited a quasilinear relationship with anion concentrations up to 100 mM. Application of 25 mM bicarbonate did not produce measurable leak currents. The leak current displayed outward rectification, which disappeared when external Na(+) was replaced by N-methyl-d-glucamine(+). More precisely, external Na(+) blocked the leak current in both directions, but its K(i) value rose rapidly when membrane potential became positive. Thus SMCT1 possesses a anionic leak current that becomes significant whenever external Na(+) concentration is reduced. The presence of this leak current may represent a second function for SMCT1 in addition to cotransporting short chain fatty acids, and future experiments will determine whether this function serves a physiological role in tissues where SMCT1 is expressed.


Assuntos
Ânions/metabolismo , Transporte Biológico , Animais , Ânions/farmacologia , Bicarbonatos/metabolismo , Ciclamatos/farmacologia , Ácidos Graxos/metabolismo , Feminino , Humanos , Potenciais da Membrana/fisiologia , Técnicas de Patch-Clamp , RNA Mensageiro/genética , Xenopus laevis
6.
Biochim Biophys Acta ; 1768(5): 1154-9, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17306760

RESUMO

Myo-inositol (MI) is involved in several important aspects of cell physiology including cell signaling and the control of intracellular osmolarity i.e. by serving as a "compatible osmolyte". Currently, three MI cotransporters have been identified: two are Na(+)-dependent (SMIT1 and SMIT2) and one is H(+)-dependent (HMIT) and predominantly expressed in the brain. The goal of this study was to characterize the expression of SMIT2 in rabbit kidney and to compare it to SMIT1. First, we quantified mRNA levels for both transporters using quantitative real-time PCR and found that SMIT1 was predominantly expressed in the medulla while SMIT2 was mainly in the cortex. This distribution of SMIT2 was confirmed on Western blots where an antibody raised against a SMIT2 epitope specifically detected a 75 kDa protein in both tissues. Characterization of MI transport in brush-border membrane vesicles (BBMV), in the presence of d-chiro-inositol and l-fucose to separately identify SMIT1 and SMIT2 activities, showed that only SMIT2 is expressed at the luminal side of proximal convoluted tubules. We thus conclude that, in the rabbit kidney, SMIT2 is predominantly expressed in the cortex where it is probably responsible for the apical transport of MI into the proximal tubule.


Assuntos
Inositol/metabolismo , Rim/metabolismo , Proteínas de Transporte de Monossacarídeos/genética , Proteínas de Transporte de Monossacarídeos/metabolismo , Sódio/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Western Blotting , Regulação da Expressão Gênica/efeitos dos fármacos , Rim/efeitos dos fármacos , Cinética , Masculino , Metilglucosídeos/farmacologia , Microvilosidades/efeitos dos fármacos , Coelhos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Vesículas Transportadoras/efeitos dos fármacos
7.
Mol Genet Metab ; 95(1-2): 81-95, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18675571

RESUMO

Ablation of the murine Slc5a3 gene results in severe myo-inositol (Ins) deficiency and congenital central apnea due to abnormal respiratory rhythmogenesis. The lethal knockout phenotype may be rescued by supplementing the maternal drinking water with 1% Ins. In order to test the hypothesis that Ins deficiency leads to inositide deficiencies, which are corrected by prenatal treatment, we measured the effects of Ins rescue on Ins, phosphatidylinositol (PtdIns) and myo-inositol polyphosphate levels in brains of E18.5 knockout fetuses. As the Slc5a3 gene structure is unique in the sodium/solute cotransporter (SLC5) family, and exon 1 is shared with the mitochondrial ribosomal protein subunit 6 (Mrps6) gene, we also sought to determine whether expression of its cognate Mrps6 gene is abnormal in knockout fetuses. The mean level of Ins was increased by 92% in brains of rescued Slc5a3 knockout fetuses (0.48 versus 0.25 nmol/mg), but was still greatly reduced in comparison to wildtype (6.97 nmol/mg). The PtdIns, InsP(5) and InsP(6) levels were normal without treatment. Mrps6 gene expression was unaffected in the E18.5 knockout fetuses. This enigmatic model is not associated with neonatal PtdIns deficiency and rescue of the phenotype may be accomplished without restoration of Ins. The biochemical mechanism that both uniformly leads to death and allows for Ins rescue remains unknown. In conclusion, in neonatal brain tissue, Mrps6 gene expression may not be contingent on function of its embedded Slc5a3 gene, while inositide deficiency may not be the mechanism of lethal apnea in null Slc5a3 mice.


Assuntos
Apneia/metabolismo , Encéfalo/metabolismo , Expressão Gênica , Inositol/metabolismo , Proteínas Mitocondriais/metabolismo , Fosfatidilinositóis/metabolismo , Proteínas Ribossômicas/metabolismo , Simportadores/deficiência , Sequência de Aminoácidos , Animais , Apneia/embriologia , Apneia/genética , Apneia/patologia , Encéfalo/embriologia , Encéfalo/patologia , Humanos , Camundongos , Camundongos Knockout , Proteínas Mitocondriais/química , Proteínas Mitocondriais/genética , Dados de Sequência Molecular , Fenótipo , Filogenia , Proteínas Ribossômicas/química , Proteínas Ribossômicas/genética , Alinhamento de Sequência , Medula Espinal , Simportadores/química , Simportadores/genética , Vertebrados/classificação , Vertebrados/genética
8.
Biochim Biophys Acta ; 1712(2): 173-84, 2005 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-15904891

RESUMO

The accessibility of the hydrophilic loop between putative transmembrane segments XIII and XIV of the Na+/glucose cotransporter (SGLT1) was studied in Xenopus oocytes, using the substituted cysteine accessibility method (SCAM) and fluorescent labelling. Fifteen cysteine mutants between positions 565 and 664 yielded cotransport currents of similar amplitude than the wild-type SGLT1 (wtSGLT1). Extracellular, membrane-impermeant MTSES(-) and MTSET(+) had no effect on either cotransport or Na+ leak currents of wtSGLT1 but 9 mutants were affected by MTSES and/or MTSET. We also performed fluorescent labelling on SGLT1 mutants, using tetramethylrhodamine-5-maleimide and showed that positions 586, 588 and 624 were accessible. As amino acids 604 to 610 in SGLT1 have been proposed to form part of a phlorizin (Pz) binding site, we measured the K(i)(Pz) and K(m)(alphaMG) for wtSGLT1 and for cysteine mutants at positions 588, 605-608 and 625. Although mutants A605C, Y606C and D607C had slightly higher K(i)(Pz) values than wtSGLT1 with minimal changes in K(m)((alpha)MG), the effects were modest and do not support the original hypothesis. We conclude that the large, hydrophilic loop near the carboxyl terminus of SGLT1 is thus accessible to the external solution but does not appear to play a major part in the binding of phlorizin.


Assuntos
Membrana Celular/metabolismo , Corantes Fluorescentes/farmacologia , Glicoproteínas de Membrana/química , Proteínas de Transporte de Monossacarídeos/química , Sequência de Aminoácidos , Animais , Transporte Biológico , Cisteína/química , DNA Complementar/metabolismo , Eletrofisiologia , Glucose/metabolismo , Humanos , Cinética , Microscopia de Fluorescência , Modelos Biológicos , Dados de Sequência Molecular , Mutação , Oócitos/metabolismo , Peptídeos/química , Florizina/química , Conformação Proteica , Isoformas de Proteínas , Estrutura Terciária de Proteína , Rodaminas/farmacologia , Homologia de Sequência de Aminoácidos , Sódio/química , Sódio/metabolismo , Transportador 1 de Glucose-Sódio , Xenopus laevis/metabolismo
9.
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
10.
Am J Physiol Cell Physiol ; 295(3): C791-9, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18650262

RESUMO

Myo-inositol (MI) is a compatible osmolyte used by cells to compensate for changes in the osmolarity of their surrounding milieu. In kidney, the basolateral Na(+)-MI cotransporter (SMIT1) and apical SMIT2 proteins are homologous cotransporters responsible for cellular uptake of MI. It has been shown in the Madin-Darby canine kidney (MDCK) cell line that SMIT1 expression was under the control of the tonicity-sensitive transcription factor, tonicity-responsive enhancer binding protein (TonEBP). We used an MDCK cell line stably transfected with SMIT2 to determine whether variations in external osmolarity could also affect SMIT2 function. Hyperosmotic conditions (+200 mosM raffinose or NaCl but not urea) generated an increase in SMIT2-specific MI uptake by three- to ninefold in a process that required protein synthesis. Using quantitative RT-PCR, we have determined that hyperosmotic conditions augment both the endogenous SMIT1 and the transfected SMIT2 mRNAs. Transport activities for both SMIT1 and SMIT2 exhibited differences in their respective induction profiles for both their sensitivities to raffinose, as well as in their time course of induction. Application of MG-132, which inhibits nuclear translocation of TonEBP, showed that the effect of osmolarity on transfected SMIT2 was unrelated to TonEBP, unlike the effect observed with SMIT1. Inhibition studies involving the hyperosmolarity-related MAPK suggested that p38 and JNK play a role in the induction of SMIT2. Further studies have shown that hyperosmolarity also upregulates another transfected transporter (Na(+)-glucose), as well as several endogenously expressed transport systems. This study shows that hyperosmolarity can stimulate transport in a TonEBP-independent manner by increasing the amount of mRNA derived from an exogenous DNA segment.


Assuntos
Fosfatos de Inositol/metabolismo , Sódio/metabolismo , Simportadores/metabolismo , Animais , Linhagem Celular , Inibidores de Cisteína Proteinase/farmacologia , Cães , Soluções Hipertônicas/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Cinética , Fatores de Transcrição NFATC/antagonistas & inibidores , Fatores de Transcrição NFATC/metabolismo , Pressão Osmótica , Isoformas de Proteínas , Inibidores de Proteínas Quinases/farmacologia , RNA Mensageiro/metabolismo , Rafinose/metabolismo , Solução Salina Hipertônica/metabolismo , Simportadores/efeitos dos fármacos , Simportadores/genética , Transfecção , Regulação para Cima , Ureia/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA