Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Nat Rev Nephrol ; 15(8): 482-500, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31168066

RESUMO

Over the past 25 years, successive cloning of SLC34A1, SLC34A2 and SLC34A3, which encode the sodium-dependent inorganic phosphate (Pi) cotransport proteins 2a-2c, has facilitated the identification of molecular mechanisms that underlie the regulation of renal and intestinal Pi transport. Pi and various hormones, including parathyroid hormone and phosphatonins, such as fibroblast growth factor 23, regulate the activity of these Pi transporters through transcriptional, translational and post-translational mechanisms involving interactions with PDZ domain-containing proteins, lipid microdomains and acute trafficking of the transporters via endocytosis and exocytosis. In humans and rodents, mutations in any of the three transporters lead to dysregulation of epithelial Pi transport with effects on serum Pi levels and can cause cardiovascular and musculoskeletal damage, illustrating the importance of these transporters in the maintenance of local and systemic Pi homeostasis. Functional and structural studies have provided insights into the mechanism by which these proteins transport Pi, whereas in vivo and ex vivo cell culture studies have identified several small molecules that can modify their transport function. These small molecules represent potential new drugs to help maintain Pi homeostasis in patients with chronic kidney disease - a condition that is associated with hyperphosphataemia and severe cardiovascular and skeletal consequences.


Assuntos
Proteínas de Transporte de Fosfato/metabolismo , Fosfatos/metabolismo , Animais , Humanos , Rim/metabolismo , Nefropatias/metabolismo
3.
J Bone Miner Res ; 30(10): 1925-37, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25827490

RESUMO

The Na(+) -dependent phosphate-cotransporter NaPi-IIb (SLC34A2) is widely expressed, with intestine, lung, and testis among the organs with highest levels of mRNA abundance. In mice, the intestinal expression of NaPi-IIb is restricted to the ileum, where the cotransporter localizes specifically at the brush border membrane (BBM) and mediates the active transport of inorganic phosphate (Pi). Constitutive full ablation of NaPi-IIb is embryonically lethal whereas the global but inducible removal of the transporter in young mice leads to intestinal loss of Pi and lung calcifications. Here we report the generation of a constitutive but intestinal-specific NaPi-IIb/Slc34a2-deficient mouse model. Constitutive intestinal ablation of NaPi-IIb results in viable pups with normal growth. Homozygous mice are characterized by fecal wasting of Pi and complete absence of Na/Pi cotransport activity in BBM vesicles (BBMVs) isolated from ileum. In contrast, the urinary excretion of Pi is reduced in these animals. The plasma levels of Pi are similar in wild-type and NaPi-IIb-deficient mice. In females, the reduced phosphaturia associates with higher expression of NaPi-IIa and higher Na/Pi cotransport activity in renal BBMVs, as well as with reduced plasma levels of intact FGF-23. A similar trend is found in males. Thus, NaPi-IIb is the only luminal Na(+) -dependent Pi transporter in the murine ileum and its absence is fully compensated for in adult females by a mechanism involving the bone-kidney axis. The contribution of this mechanism to the adaptive response is less apparent in adult males.


Assuntos
Íleo/metabolismo , Rim/metabolismo , Microvilosidades/metabolismo , Fosfatos/sangue , Fosfatos/urina , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/metabolismo , Animais , Transporte Biológico Ativo/genética , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/sangue , Hipofosfatemia Familiar/metabolismo , Íleo/patologia , Rim/patologia , Masculino , Camundongos , Camundongos Knockout , Microvilosidades/genética , Microvilosidades/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/genética
4.
Am J Physiol Renal Physiol ; 306(8): F833-43, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24553430

RESUMO

The proximal renal epithelia express three different Na-dependent inorganic phosphate (Pi) cotransporters: NaPi-IIa/SLC34A1, NaPi-IIc/SLC34A3, and PiT2/SLC20A2. Constitutive mouse knockout models of NaPi-IIa and NaPi-IIc suggested that NaPi-IIa mediates the bulk of renal reabsorption of Pi whereas the contribution of NaPi-IIc to this process is minor and probably restricted to young mice. However, many reports indicate that mutations of NaPi-IIc in humans lead to hereditary hypophosphatemic rickets with hypercalciuria (HHRH). Here, we report the generation of a kidney-specific and inducible NaPi-IIc-deficient mouse model based on the loxP-Cre system. We found that the specific removal of the cotransporter from the kidneys of young mice does not impair the capacity of the renal epithelia to transport Pi. Moreover, the levels of Pi in plasma and urine as well as the circulating levels of parathyroid hormone, FGF-23, and vitamin D3 remained unchanged. These findings are in agreement with the data obtained with the constitutive knockout model and suggest that, under steady-state conditions of normal dietary Pi, NaPi-IIc is not an essential Na-Pi cotransporter in murine kidneys. However, and unlike the constitutive mutants, the kidney-specific depletion of NaPi-IIc does not result in alteration of the homeostasis of calcium. This suggests that the calcium-related phenotype observed in constitutive knockout mice may not be related to inactivation of the cotransporter in kidney.


Assuntos
Cálcio/metabolismo , Fosfatos/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIc/genética , Animais , Calcitriol/metabolismo , Doxiciclina/farmacologia , Raquitismo Hipofosfatêmico Familiar/fisiopatologia , Fator de Crescimento de Fibroblastos 23 , Homeostase/efeitos dos fármacos , Rim/metabolismo , Masculino , Camundongos , Camundongos Knockout , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIc/deficiência
5.
Cell Physiol Biochem ; 32(1): 187-99, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23899881

RESUMO

BACKGROUND/AIMS: Members of the SLC20 cotransporter family (PiT-1, PiT-2) are ubiquitously expressed in mammalian tissue and are thought to perform housekeeping functions for intracellular Pi homeostasis as well as being implicated in vascular calcification and renal Pi reabsorption. The aims of this study were to investigate the topology of a linker region in PiT-1 between the predicted 2(nd) and 3(rd) transmembrane domains and to investigate the functional consequences of cysteine substitutions in this region. METHODS: Cysteines were substituted at 18 sites in the Xenopus PiT-1 isoform and the mutants were expressed in Xenopus laevis oocytes. Transport function of the mutants was investigated by (32)P tracer or two electrode voltage clamp before and after thiol modification of the novel Cys. RESULTS: Exposure to the thiol reactive reagent resulted in diminished transport function for 7 mutants. The apparent accessibility of 5 of the mutated sites, estimated from the rate of functional thiol modification, was site-dependent. Cysteine substitution at some sites also altered the apparent affinity for Pi and cation (Na(+)/Li(+)) and substrate (phosphate/arsenate) selectivity, further underscoring the importance of this linker in defining PiT-1 transport characteristics. CONCLUSIONS: The external accessibility of a linker in PiT-1 was confirmed and sites were identified that determine substrate selectivity and transport function.


Assuntos
Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/metabolismo , Sequência de Aminoácidos , Animais , Transporte Biológico , Cisteína/química , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Oócitos/fisiologia , Técnicas de Patch-Clamp , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/química , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/genética , Especificidade por Substrato , Xenopus laevis/crescimento & desenvolvimento , Xenopus laevis/metabolismo
6.
Pflugers Arch ; 465(11): 1557-72, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23708836

RESUMO

Renal reabsorption of inorganic phosphate (Pi) is mediated by the phosphate transporters NaPi-IIa, NaPi-IIc, and Pit-2 in the proximal tubule brush border membrane (BBM). Dietary Pi intake regulates these transporters; however, the contribution of the specific isoforms to the rapid and slow phase is not fully clarified. Moreover, the regulation of PTH and FGF23, two major phosphaturic hormones, during the adaptive phase has not been correlated. C57/BL6 and NaPi-IIa(-/-) mice received 5 days either 1.2 % (HPD) or 0.1 % (LPD) Pi-containing diets. Thereafter, some mice were acutely switched to LPD or HPD. Plasma Pi concentrations were similar under chronic diets, but lower when mice were acutely switched to LPD. Urinary Pi excretion was similar in C57/BL6 and NaPi-IIa(-/-) mice under HPD. During chronic LPD, NaPi-IIa(-/-) mice lost phosphate in urine compensated by higher intestinal Pi absorption. During the acute HPD-to-LPD switch, NaPi-IIa(-/-) mice exhibited a delayed decrease in urinary Pi excretion. PTH was acutely regulated by low dietary Pi intake. FGF23 did not respond to low Pi intake within 8 h whereas the phospho-adaptator protein FRS2α necessary for FGF-receptor cell signaling was downregulated. BBM Pi transport activity and NaPi-IIa but not NaPi-IIc and Pit-2 abundance acutely adapted to diets in C57/BL6 mice. In NaPi-IIa(-/-), Pi transport activity was low and did not adapt. Thus, NaPi-IIa mediates the fast adaptation to Pi intake and is upregulated during the adaptation to low Pi despite persistently high FGF23 levels. The sensitivity to FGF23 may be regulated by adapting FRS2α abundance and phosphorylation.


Assuntos
Adaptação Fisiológica , Fatores de Crescimento de Fibroblastos/metabolismo , Túbulos Renais Proximais/metabolismo , Proteínas de Membrana/metabolismo , Fósforo na Dieta/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Fator de Crescimento de Fibroblastos 23 , Absorção Intestinal , Túbulos Renais Proximais/fisiologia , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fósforo na Dieta/sangue , Fósforo na Dieta/urina , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/metabolismo
7.
Mol Aspects Med ; 34(2-3): 386-95, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23506879

RESUMO

Transport of inorganic phosphate (Pi) across the plasma membrane is essential for normal cellular function. Members of two families of SLC proteins (SLC20 and SLC34) act as Na(+)-dependent, secondary-active cotransporters to transport Pi across cell membranes. The SLC34 proteins are expressed in specific organs important for Pi homeostasis: NaPi-IIa (SLC34A1) and NaPi-IIc (SLC34A3) fulfill essential roles in Pi reabsorption in the kidney proximal tubule and NaPi-IIb (SLC34A2) mediates Pi absorption in the gut. The SLC20 proteins, PiT-1 (SLC20A1), PiT-2 (SLC20A2) are expressed ubiquitously in all tissues and although generally considered as "housekeeping" transport proteins, the discovery of tissue-specific activity, regulatory pathways and gene-related pathophysiologies, is redefining their importance. This review summarizes our current knowledge of SLC20 and SLC34 proteins in terms of their basic molecular characteristics, physiological roles, known pathophysiology and pharmacology.


Assuntos
Homeostase/fisiologia , Modelos Moleculares , Família Multigênica/genética , Conformação Proteica , Proteínas Cotransportadoras de Sódio-Fosfato/genética , Proteínas Cotransportadoras de Sódio-Fosfato/fisiologia , Foscarnet/farmacologia , Humanos , Cinética , Modelos Biológicos , Proteínas Cotransportadoras de Sódio-Fosfato/metabolismo
8.
Curr Top Membr ; 70: 313-56, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23177991

RESUMO

Transport of inorganic phosphate (P(i)) is mediated by proteins belonging to two solute carrier families (SLC20 and SLC34). Members of both families transport P(i) using the electrochemical gradient for Na(+). The role of the SLC34 members as essential players in mammalian P(i) homeostasis is well established, whereas that of SLC20 proteins is less well defined. The SLC34 family comprises the following three isoforms that preferentially cotransport divalent P(i) and are expressed in epithelial tissue: the renal NaPi-IIa and NaPi-IIc are responsible for reabsorbing P(i) in the proximal tubule, whereas NaPi-IIb is more ubiquitously expressed, including the small intestine, where it mediates dietary P(i) absorption. The SLC20 family comprises two members (PiT-1, PiT-2) that preferentially cotransport monovalent P(i) and are expressed in epithelial as well as nonepithelial tissue. The transport kinetics of members of both families have been characterized in detail using heterologous expression in Xenopus oocytes. For the electrogenic NaPi-IIa/b, and PiT-1,-2, conventional electrophysiological techniques together with radiotracer methods have been applied, as well as time-resolved fluorometric measurements that allow new insights into local conformational changes of the protein during the cotransport cycle. For the electroneutral NaPi-IIc, conventional tracer uptake and fluorometry have been used to elucidate its transport properties. The 3-D structures of these proteins remain unresolved and structure-function studies have so far concentrated on defining the topology and identifying sites of functional importance.


Assuntos
Fosfatos/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/química , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/química , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/metabolismo , Animais , Cátions/metabolismo , Túbulos Renais Proximais/metabolismo , Cinética , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Relação Estrutura-Atividade , Especificidade por Substrato
10.
Am J Physiol Cell Physiol ; 302(3): C539-54, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22075694

RESUMO

Type IIa/b Na(+)-coupled inorganic phosphate cotransporters (NaPi-IIa/b) are considered to be exclusively Na(+) dependent. Here we show that Li(+) can substitute for Na(+) as a driving cation. We expressed NaPi-IIa/b in Xenopus laevis oocytes and performed two-electrode voltage-clamp electrophysiology and uptake assays to investigate the effect of external Li(+) on their kinetics. Replacement of 50% external Na(+) with Li(+) reduced the maximum transport rate and the rate-limiting plateau of the P(i)-induced current began at less hyperpolarizing potentials. Simultaneous electrophysiology and (22)Na uptake on single oocytes revealed that Li(+) ions can substitute for at least one of the three Na(+) ions necessary for cotransport. Presteady-state assays indicated that Li(+) ions alone interact with the empty carrier; however, the total charge displaced was 70% of that with Na(+) alone, or when 50% of the Na(+) was replaced by Li(+). If Na(+) and Li(+) were both present, the midpoint potential of the steady-state charge distribution was shifted towards depolarizing potentials. The charge movement in the presence of Li(+) alone reflected the interaction of one Li(+) ion, in contrast to 2 Na(+) ions when only Na was present. We propose an ordered binding scheme for cotransport in which Li(+) competes with Na(+) to occupy the putative first cation interaction site, followed by the cooperative binding of one Na(+) ion, one divalent P(i) anion, and a third Na(+) ion to complete the carrier loading. With Li(+) bound, the kinetics of subsequent partial reactions were significantly altered. Kinetic simulations of this scheme support our experimental data.


Assuntos
Lítio/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/metabolismo , Sódio/metabolismo , Animais , Transporte Biológico , Potenciais da Membrana , Oócitos/citologia , Oócitos/metabolismo , Técnicas de Patch-Clamp , Fosfatos/metabolismo , Xenopus laevis
11.
Pflugers Arch ; 461(6): 645-63, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21384128

RESUMO

A common structural feature characterises sodium-coupled inorganic phosphate cotransporters of the SLC34 family (NaPi-IIa/b/c): a pair of inverted regions in the N- and C-terminal halves of the protein. These regions are hypothesised to contain re-entrant domains that associate to allow alternating access of the substrates from either side of the membrane. To investigate if these domains interact during the NaPi-II transport cycle, we introduced novel cysteines at three functionally important sites associated with the predicted re-entrant domains of the flounder NaPi-IIb for the purpose of fluorescent labelling and cross-linking. Single and double mutants were expressed in Xenopus oocytes and their function analysed using electrophysiological and real-time fluorometric assays. The substitution at the cytosolic end of the first re-entrant domain induced a large hyperpolarizing shift in the voltage dependence of steady-state and presteady-state kinetics, whereas the two substitutions at the external face were less critical. By using Cu-phenanthroline to induce disulfide bridge formation, we observed a loss of transport activity that depended on the presence of sodium in the incubation medium. This suggested that external sodium increased the probability of NaPi-IIb occupying a conformation that favours interaction between sites in the re-entrant domains. Furthermore, voltage-dependent fluorescence data supported the hypothesis that a localised interaction between the two domains occurs that depends on the membrane potential and substrate present: we found that the fluorescence intensity reported by a labelled cysteine in one domain was dependent on the side chain substituted at a functionally critical site in the opposed domain.


Assuntos
Proteínas Cotransportadoras de Sódio-Fosfato/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Sítios de Ligação , Cisteína/metabolismo , Linguado , Dados de Sequência Molecular , Oócitos/metabolismo , Técnicas de Patch-Clamp , Estrutura Terciária de Proteína , Proteínas Cotransportadoras de Sódio-Fosfato/química , Proteínas Cotransportadoras de Sódio-Fosfato/genética , Xenopus laevis
12.
J Nephrol ; 23 Suppl 16: S145-51, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21170872

RESUMO

In kidneys of mammals, filtered phosphate ions (Pi) are reabsorbed along the proximal tubules. Transcellular transport of phosphate is initiated by several apically localized sodium-dependent Pi cotransporters (Na/Pi-cotransporters) that belong to the SLC 20 (SLC20A2) and 34 (SLC34A1, SLC34A3) families. Apical abundance of these Na/Pi-cotransporters is adjusted by numerous hormones/phosphatonins and metabolic factors in order to adjust the extent of renal Pi reabsorption according to body needs. Acute hormonal regulation of Pi reabsorption occurs mainly by a change of the abundance of SLC34A1 (NaPi-IIa) via modulation of the interaction of NaPi-IIa with the PDZ-protein NHERF1.


Assuntos
Rim/metabolismo , Fosfatos/metabolismo , Absorção , Animais , Dopamina/fisiologia , Humanos , Concentração de Íons de Hidrogênio , Transporte de Íons , Hormônio Paratireóideo/fisiologia , Fosfatos/administração & dosagem , Fosfoproteínas/fisiologia , Trocadores de Sódio-Hidrogênio/fisiologia , Proteínas Cotransportadoras de Sódio-Fosfato/fisiologia
14.
Urol Res ; 38(4): 271-6, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20665015

RESUMO

Control of phosphate (P(i)) homeostasis is essential for many biologic functions and inappropriate low levels of P(i) in plasma have been suggested to associate with several pathological states, including renal stone formation and stone recurrence. P(i) homeostasis is achieved mainly by adjusting the renal reabsorption of P(i) to the body's requirements. This task is performed to a major extent by the Na/Pi cotransporter NaPi-IIa that is specifically expressed in the brush border membrane of renal proximal tubules. While the presence of tight junctions in epithelial cells prevents the diffusion and mixing of the apical and basolateral components, the location of a protein within a particular membrane subdomain (i.e., the presence of NaPi-IIa at the tip of the apical microvilli) often requires its association with scaffolding elements which directly or indirectly connect the protein with the underlying cellular cytoskeleton. NaPi-IIa interacts with the four members of the Na(+)/H(+) exchanger regulatory factor family as well as with the GABA(A)-receptor associated protein . Here we will discuss the most relevant findings regarding the role of these proteins on the expression and regulation of the cotransporter, as well as the impact that their absence has in P(i) homeostasis.


Assuntos
Rim/metabolismo , Fosfatos/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/fisiologia , Absorção , Animais , Homeostase , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/metabolismo , Modelos Biológicos , Domínios PDZ/fisiologia , Fosfoproteínas/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo
15.
Pflugers Arch ; 460(3): 677-87, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20526720

RESUMO

Renal phosphate reabsorption across the brush border membrane (BBM) in the proximal tubule is mediated by at least three transporters, NaPi-IIa (SLC34A1), NaPi-IIc (SLC34A3), and Pit-2 (SLC20A2). Parathyroid hormone (PTH) is a potent phosphaturic factor exerting an acute and chronic reduction in proximal tubule phosphate reabsorption. PTH acutely induces NaPi-IIa internalization from the BBM and lysosomal degradation, but its effects on NaPi-IIc and Pit-2 are unknown. In rats adapted to low phosphate diet, acute (30 and 60 min) application of PTH decreased BBM phosphate transport rates both in the absence and the presence of phosphonoformic acid, an inhibitor of SLC34 but not SLC20 transporters. Immunohistochemistry showed NaPi-IIa expression in the S1 to the S3 segment of superficial and juxtamedullary nephrons; NaPi-IIc was only detectable in S1 segments and Pit-2 in S1 and weakly in S2 segments of superficial and juxtamedullary nephrons. PTH reduced NaPi-IIa staining in the BBM with increased intracellular and lysosomal appearance. NaPi-IIa internalization was most prominent in S1 segments of superficial nephrons. We did not detect changes in NaPi-IIc and Pit-2 staining over this time period. Blockade of lysosomal protein degradation with leupeptin revealed NaPi-IIa accumulation in lysosomes, but no lysosomal staining for NaPi-IIc or Pit-2 could be detected. Immunoblotting of BBM confirmed the reduction in NaPi-IIa abundance and the absence of any effect on NaPi-IIc expression. Pit-2 protein abundance was also significantly reduced by PTH. Thus, function and expression of BBM phosphate cotransporters are differentially regulated allowing for fine-tuning of renal phosphate reabsorption.


Assuntos
Rim/metabolismo , Hormônio Paratireóideo/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato/metabolismo , Animais , Rim/ultraestrutura , Lisossomos/metabolismo , Masculino , Microvilosidades/metabolismo , Fosfatos/metabolismo , Ratos , Ratos Wistar , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIc/metabolismo
16.
Pflugers Arch ; 460(1): 207-17, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20354864

RESUMO

We have recently shown that the abundance of the renal sodium (Na)/inorganic phosphate (Pi) cotransporter NaPi-IIa is increased in the absence of the GABA(A) receptor-associated protein (GABARAP). Accordingly, GABARAP-deficient mice have a reduced urinary excretion of Pi. However, their circulating levels of Pi do not differ from wild-type animals, suggesting the presence of a compensatory mechanism responsible for keeping serum Pi values constant. Here, we aimed first to identify the molecular basis of this compensation by analyzing the expression of Na/Pi cotransporters known to be expressed in the kidney and intestine. We found that, in the kidney, the upregulation of NaPi-IIa is not accompanied by changes on the expression of either NaPi-IIc or PiT2, the other cotransporters known to participate in renal Pi reabsorption. In contrast, the intestinal expression of NaPi-IIb is downregulated in mutant animals, suggesting that a reduced intestinal absorption of Pi could contribute to maintain a normophosphatemic status despite the increased renal retention. The second goal of this work was to study whether the alterations on the expression of NaPi-IIa induced by chronic dietary Pi are impaired in the absence of GABARAP. Our data indicate that, in response to high Pi diets, GABARAP-deficient mice downregulate the expression of NaPi-IIa to levels comparable to those seen in wild-type animals. However, in response to low Pi diets, the upregulation of NaPi-IIa is greater in the mutant mice. Thus, both the basal expression and the dietary-induced upregulation of NaPi-IIa are increased in the absence of GABARAP.


Assuntos
Proteínas do Citoesqueleto/deficiência , Íleo/metabolismo , Rim/metabolismo , Proteínas de Membrana/deficiência , Fosfatos/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/metabolismo , Sódio/metabolismo , Animais , Proteínas Reguladoras de Apoptose , Densidade Óssea , Proteínas do Citoesqueleto/genética , Fêmur/metabolismo , Regulação da Expressão Gênica , Absorção Intestinal , Vértebras Lombares/metabolismo , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Associadas aos Microtúbulos , Microvilosidades/metabolismo , Fosfatos/sangue , Fosfatos/urina , Fósforo na Dieta/metabolismo , RNA Mensageiro/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/genética
17.
J Physiol ; 587(Pt 17): 4293-307, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-19596895

RESUMO

The SLC34 solute carrier family comprises the electrogenic NaPi-IIa/b and the electroneutral NaPi-IIc, which display Na(+) : P(i) cotransport stoichiometries of 3 : 1 and 2 : 1, respectively. We previously proposed that NaPi-IIc lacks one of the three Na(+) interaction sites hypothesised for the electrogenic isoforms, but, unlike NaPi-IIa/b, its substrate binding order is undetermined. By expressing NaPi-IIc in Xenopus oocytes, isotope influx and efflux assays gave results consistent with Na(+) being the first and last substrate to bind. To further investigate substrate interactions, we applied a fluorometry-based technique that uses site-specific labelling with a fluorophore to characterize substrate-induced conformational changes. A novel Cys was introduced in the third extracellular loop of NaPi-IIc that could be labelled with a reporter fluorophore (MTS-TAMRA). Although labelling resulted in suppression of cotransport as previously reported for the electrogenic isoforms, changes in fluorescence were induced by changes in extracellular Na(+) concentration in the absence of P(i) and by changes in extracellular P(i) concentration in presence of Na(+). These data, combined with (32)P uptake data, also support a binding scheme in which Na(+) is the first substrate to interact. Moreover, the apparent P(i) affinity from fluorometry agreed with that from (32)P uptake, confirming the applicability of the fluorometric technique for kinetic studies of electroneutral carriers. Analysis of the fluorescence data showed that like the electrogenic NaPi-IIb, 2 Na(+) ions interact cooperatively with NaPi-IIc before P(i) binding, which implies that only one of these is translocated. This result provides compelling evidence that SLC34 proteins share common motifs for substrate interaction and that cotransport and substrate binding stoichiometries are not necessarily equivalent.


Assuntos
Ativação do Canal Iônico/fisiologia , Potenciais da Membrana/fisiologia , Oócitos/fisiologia , Proteínas Cotransportadoras de Sódio-Fosfato/classificação , Proteínas Cotransportadoras de Sódio-Fosfato/fisiologia , Sódio/metabolismo , Animais , Células Cultivadas , Xenopus laevis
18.
Am J Physiol Renal Physiol ; 296(5): F1118-28, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19225049

RESUMO

Renal reabsorption of inorganic phosphate (P(i)) is mainly mediated by the Na(+)-dependent P(i)-cotransporter NaPi-IIa that is expressed in the brush-border membrane (BBM) of renal proximal tubules. Regulation and apical expression of NaPi-IIa are known to depend on a network of interacting proteins. Most of the interacting partners identified so far associate with the COOH-terminal PDZ-binding motif (TRL) of NaPi-IIa. In this study GABA(A) receptor-associated protein (GABARAP) was identified as a novel interacting partner of NaPi-IIa applying a membrane yeast-two-hybrid system (MYTH 2.0) to screen a mouse kidney library with the TRL-truncated cotransporter as bait. GABARAP mRNA and protein are present in renal tubules, and the interaction of NaPi-IIa and GABARAP was confirmed by using glutathione S-transferase pulldowns from BBM and coimmunoprecipitations from transfected HEK293 cells. Amino acids 36-68 of GABARAP were identified as the determinant for the described interaction. The in vivo effects of this interaction were studied in a murine model. GABARAP(-/-) mice have reduced urinary excretion of P(i), higher Na(+)-dependent (32)P(i) uptake in BBM vesicles, and increased expression of NaPi-IIa in renal BBM compared with GABARAP(+/+) mice. The expression of Na(+)/H(+) exchanger regulatory factor (NHERF)1, an important scaffold for the apical expression of NaPi-IIa, is also increased in GABARAP(-/-) mice. The absence of GABARAP does not interfere with the regulation of the cotransporter by either parathyroid hormone or acute changes of dietary P(i) content.


Assuntos
Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Túbulos Renais Proximais/fisiologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/metabolismo , Animais , Proteínas Reguladoras de Apoptose , Linhagem Celular , Células-Tronco Embrionárias/fisiologia , Endocitose/efeitos dos fármacos , Endocitose/fisiologia , Células Epiteliais/fisiologia , Expressão Gênica/fisiologia , Biblioteca Gênica , Homeostase/fisiologia , Humanos , Rim/citologia , Túbulos Renais Proximais/citologia , Camundongos , Camundongos Mutantes , Proteínas Associadas aos Microtúbulos , Microvilosidades/fisiologia , Hormônio Paratireóideo/farmacologia , Fosfatos/metabolismo , Fosfatos/farmacologia , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fósforo na Dieta/farmacologia , RNA Mensageiro/metabolismo , Trocadores de Sódio-Hidrogênio/genética , Trocadores de Sódio-Hidrogênio/metabolismo
19.
Am J Physiol Renal Physiol ; 296(4): F691-9, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19073637

RESUMO

The principal mediators of renal phosphate (P(i)) reabsorption are the SLC34 family proteins NaPi-IIa and NaPi-IIc, localized to the proximal tubule (PT) apical membrane. Their abundance is regulated by circulatory factors and dietary P(i). Although their physiological importance has been confirmed in knockout animal studies, significant P(i) reabsorptive capacity remains, which suggests the involvement of other secondary-active P(i) transporters along the nephron. Here we show that a member of the SLC20 gene family (PiT-2) is localized to the brush-border membrane (BBM) of the PT epithelia and that its abundance, confirmed by Western blot and immunohistochemistry of rat kidney slices, is regulated by dietary P(i). In rats treated chronically on a high-P(i) (1.2%) diet, there was a marked decrease in the apparent abundance of PiT-2 protein in kidney slices compared with those from rats kept on a chronic low-P(i) (0.1%) diet. In Western blots of BBM from rats that were switched from a chronic low- to high-P(i) diet, NaPi-IIa showed rapid downregulation after 2 h; PiT-2 was also significantly downregulated at 24 h and NaPi-IIc after 48 h. For the converse dietary regime, NaPi-IIa showed adaptation within 8 h, whereas PiT-2 and NaPi-IIc showed a slower adaptive trend. Our findings suggest that PiT-2, until now considered as a ubiquitously expressed P(i) housekeeping transporter, is a novel mediator of P(i) reabsorption in the PT under conditions of acute P(i) deprivation, but with a different adaptive time course from NaPi-IIa and NaPi-IIc.


Assuntos
Membrana Celular/metabolismo , Dieta , Túbulos Renais Proximais/metabolismo , Fosfatos/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/metabolismo , Adaptação Fisiológica , Animais , Transporte Biológico , Western Blotting , Polaridade Celular , Imuno-Histoquímica , Masculino , Microvilosidades/metabolismo , Fosfatos/administração & dosagem , Fosfatos/deficiência , Ratos , Ratos Wistar , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/metabolismo , Fatores de Tempo
20.
Channels (Austin) ; 2(5): 346-57, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18989094

RESUMO

Na(+)-coupled phosphate cotransporters of the SLC34 gene family catalyze the movement of inorganic phosphate (P(i)) across epithelia by using the free energy of the downhill electrochemical Na(+) gradient across the luminal membrane. Electrogenic (NaPi-IIa/b) and electroneutral (NaPi-IIc) isoforms prefer divalent P(i) and show strict Na(+):P(i) stoichiometries of 3:1 and 2:1, respectively. For electrogenic cotransport, one charge is translocated per transport cycle. When NaPi-IIa or NaPi-IIb are expressed in Xenopus oocytes, application of the P(i) transport inhibitor phosphonoformic acid (PFA) blocks a leak current that is not detectable in the electroneutral isoform. In this review, we present the experimental evidence that this transport-independent leak originates from a Na(+)-dependent uniport carrier mode intrinsic to NaPi-IIa/b isoforms. Our findings, based on the characteristics of the PFA-inhibitable leak measured from wild-type and mutant constructs, can be incorporated into an alternating access class model in which the leak and cotransport modes are mutually exclusive and share common kinetic partial reactions.


Assuntos
Transporte de Íons , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/metabolismo , Animais , Humanos , Permeabilidade , Isoformas de Proteínas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA