Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Pflugers Arch ; 471(1): 109-122, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30218374

RESUMO

The solute carrier 34 (SLC34) family of membrane transporters is a major contributor to Pi homeostasis. Many factors are involved in regulating the SLC34 family. The roles of the bone mineral metabolism factors parathyroid hormone (PTH) and fibroblast growth factor 23 (FGF23) in Pi homeostasis are well studied. Intracellular Pi is thought to be involved in energy metabolism, such as ATP production. Under certain conditions of altered energy metabolism, plasma Pi concentrations are affected by the regulation of a Pi shift into cells or release from the tissues. We recently investigated the mechanism of hepatectomy-related hypophosphatemia, which is thought to involve an unknown phosphaturic factor. Hepatectomy-related hypophosphatemia is due to impaired nicotinamide adenine dinucleotide (NAD) metabolism through its effects on the SLC34 family in the liver-kidney axis. The oxidized form of NAD, NAD+, is an essential cofactor in various cellular biochemical reactions. Levels of NAD+ and its reduced form NADH vary with the availability of dietary energy and nutrients. Nicotinamide phosphoribosyltransferase (Nampt) generates a key NAD+ intermediate, nicotinamide mononucleotide, from nicotinamide and 5-phosphoribosyl 1-pyrophosphate. The liver, an important organ of NAD metabolism, is thought to release metabolic products such as nicotinamide and may control NAD metabolism in other organs. Moreover, NAD is an important regulator of the circadian rhythm. Liver-specific Nampt-deficient mice and heterozygous Nampt mice have abnormal daily plasma Pi concentration oscillations. These data indicate that NAD metabolism in the intestine, liver, and kidney is closely related to Pi metabolism through the SLC34 family. Here, we review the relationship between the SLC34 family and NAD metabolism based on our recent studies.


Assuntos
Rim/metabolismo , Fígado/metabolismo , NAD/metabolismo , Fosfatos/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/metabolismo , Animais , Ritmo Circadiano , Fator de Crescimento de Fibroblastos 23 , Homeostase , Humanos , Fosfatos/sangue
2.
Pflugers Arch ; 471(1): 123-136, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30523405

RESUMO

Inorganic phosphate (Pi) secretion from the salivary glands and dietary Pi are key Pi sources. The regulatory mechanisms of Pi homeostasis in the salivary glands are unknown. We investigated how salivary Pi concentrations are regulated by dietary Pi in mouse models. Dietary manipulation significantly changed the levels of Npt2b protein in the salivary gland ductal cells. In addition, rapid feeding on a high-Pi diet increased the saliva Pi concentrations and led to rapid endocytosis of Npt2b in the apical membranes of the duct cells. Global Npt2b± mice exhibited increased salivary Pi concentrations and intestine-specific deletion of Npt2b after high Pi loading increased the salivary Pi concentrations. These findings indicate that Npt2b levels in the salivary glands affect the salivary Pi concentration and are regulated by dietary Pi. Intestinal Npt2b levels might also affect salivary Pi concentrations as well as renal Pi excretion. These findings suggest Pi is endogenously recycled by salivary Pi secretion, intestinal Pi absorption, and renal Pi excretion.


Assuntos
Adaptação Fisiológica , Mucosa Intestinal/metabolismo , Rim/metabolismo , Fósforo na Dieta/metabolismo , Glândulas Salivares/metabolismo , Animais , Absorção Intestinal , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosfatos/metabolismo , Eliminação Renal , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/metabolismo
3.
Clin Exp Nephrol ; 23(7): 898-907, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30895530

RESUMO

BACKGROUND: Injection of parathyroid hormone (PTH) rapidly stimulates renal Pi excretion, in part by downregulating NaPi-IIa (Npt2a/SLC34A1) and NaPi-IIc (Npt2c/SLC34A3) transporters. The mechanisms underlying the effects of PTH on NaPi-IIc are not fully elucidated. METHODS: We analyzed the effect of PTH on inorganic phosphate (Pi) reabsorption in Npt2a-/- mice to eliminate the influence of Npt2a on renal Pi reabsorption. In opossum kidney (OK) cells and Xenopus oocytes, we investigated the effect of NaPi-IIc transporter phosphorylation. Studies of mice with mutations of NaPi-IIc protein in which serine and threonine were replaced with either alanine (A), which prevents phosphorylation, or aspartic acid (D), which mimics the charged state of phosphorylated NaPi-IIc, were also performed to evaluate the involvement of phosphorylation in the regulation of transport function. RESULTS: The Npt2a-/- experiments showed that PTH administration rapidly inactivated NaPi-IIc function in the apical membrane of proximal tubular cells. Analysis of mutant proteins (S71, S138, T151, S174, T583) at putative protein kinase C sites, revealed that S138 markedly suppressed the function and cellular expression of mouse NaPi-IIc in Xenopus oocytes and OK cells. In addition, 138D had a short half-life compared with wild-type protein. CONCLUSIONS: The present study suggests that acute regulation of NaPi-IIc protein by PTH is involved in the inactivation of Na+-dependent Pi cotransporter activity and that phosphorylation of the transporter is involved in the rapid modification.


Assuntos
Túbulos Renais Proximais/efeitos dos fármacos , Hormônio Paratireóideo/farmacologia , Fragmentos de Peptídeos/farmacologia , Fosfatos/metabolismo , Proteína Quinase C/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Reabsorção Renal/efeitos dos fármacos , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIc/metabolismo , Animais , Linhagem Celular , Feminino , Túbulos Renais Proximais/metabolismo , Masculino , Camundongos Knockout , Gambás , Fosforilação , Estabilidade Proteica , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/deficiência , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIc/genética , Fatores de Tempo , Xenopus
4.
Clin Exp Nephrol ; 23(3): 313-324, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30317447

RESUMO

BACKGROUND: The role of Na+-dependent inorganic phosphate (Pi) transporters in the human kidney is not fully clarified. Hereditary hypophosphatemic rickets with hypercalciuria (HHRH) is caused by loss-of-function mutations in the IIc Na+-dependent Pi transporter (NPT2c/Npt2c/NaPi-IIc) gene. Another Na+-dependent type II transporter, (NPT2A/Npt2a/NaPi-IIa), is also important for renal Pi reabsorption in humans. In mice, Npt2c deletion does not lead to hypophosphatemia and rickets because Npt2a compensates for the impaired Pi reabsorption. To clarify the differences between mouse and human, we investigated the relation between NaPi-IIa and NaPi-IIc functions in opossum kidney (OK) cells. METHODS: We cloned NaPi-IIc from OK cells and created opossum NaPi-IIc (oNaPi-IIc) antibodies. We used oNaPi-IIc small interference (si)RNA and investigated the role of NaPi-IIc in Pi transport in OK cells. RESULTS: We cloned opossum kidney NaPi-IIc cDNAs encoding 622 amino acid proteins (variant1) and examined their pH- and sodium-dependency. The antibodies reacted specifically with 75-kDa and 150-kDa protein bands, and the siRNA of NaPi-IIc markedly suppressed endogenous oNaPi-IIc in OK cells. Treatment with siRNA significantly suppressed the expression of NaPi-4 (NaPi-IIa) protein and mRNA. oNaPi-IIc siRNA also suppressed Na+/H+ exchanger regulatory factor 1 expression in OK cells. CONCLUSION: These findings suggest that NaPi-IIc is important for the expression of NaPi-IIa (NaPi-4) protein in OK cells. Suppression of Npt2c may downregulate Npt2a function in HHRH patients.


Assuntos
Rim/metabolismo , Proteínas de Transporte de Fosfato/fisiologia , Fosfatos/metabolismo , Animais , Células Cultivadas , Raquitismo Hipofosfatêmico Familiar/etiologia , Humanos , Hipercalciúria/etiologia , Camundongos , Gambás , RNA Interferente Pequeno/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIc/fisiologia , Xenopus laevis
5.
Kidney Int ; 93(5): 1073-1085, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29398136

RESUMO

Circulating inorganic phosphate exhibits a remarkable daily oscillation based on food intake. In humans and rodents, the daily oscillation in response to food intake may be coordinated to control the intestinal absorption, renal excretion, cellular shifts, and extracellular concentration of inorganic phosphate. However, mechanisms regulating the resulting oscillation are unknown. Here we investigated the roles of the sodium phosphate cotransporter SLC34 (Npt2) family and nicotinamide phosphoribosyltransferase (Nampt) in the daily oscillation of plasma inorganic phosphate levels. First, it is roughly linked to urinary inorganic phosphate excretion. Second, expression of renal Npt2a and Npt2c, and intestinal Npt2b proteins also exhibit a dynamic daily oscillation. Analyses of Npt2a, Npt2b, and Npt2c knockout mice revealed the importance of renal inorganic phosphate reabsorption and cellular inorganic phosphate shifts in the daily oscillation. Third, experiments in which nicotinamide and a specific Nampt inhibitor (FK866) were administered in the active and rest phases revealed that the Nampt/NAD+ system is involved in renal inorganic phosphate excretion. Additionally, for cellular shifts, liver-specific Nampt deletion disturbed the daily oscillation of plasma phosphate during the rest but not the active phase. In systemic Nampt+/- mice, NAD levels were significantly reduced in the liver, kidney, and intestine, and the daily oscillation (active and rest phases) of the plasma phosphate concentration was attenuated. Thus, the Nampt/NAD+ system for Npt2 regulation and cellular shifts to tissues such as the liver play an important role in generating daily oscillation of plasma inorganic phosphate levels.


Assuntos
Ritmo Circadiano , Citocinas/metabolismo , Nicotinamida Fosforribosiltransferase/metabolismo , Fosfatos/sangue , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIc/metabolismo , Animais , Biomarcadores/sangue , Biomarcadores/urina , Citocinas/antagonistas & inibidores , Citocinas/deficiência , Citocinas/genética , Inibidores Enzimáticos/farmacologia , Feminino , Intestinos/enzimologia , Rim/enzimologia , Fígado/enzimologia , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , NAD/metabolismo , Nicotinamida Fosforribosiltransferase/antagonistas & inibidores , Nicotinamida Fosforribosiltransferase/deficiência , Nicotinamida Fosforribosiltransferase/genética , Fosfatos/urina , Eliminação Renal , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/deficiência , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/deficiência , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIc/deficiência , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIc/genética , Fatores de Tempo
6.
Kidney Blood Press Res ; 43(5): 1409-1424, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30212831

RESUMO

BACKGROUND/AIMS: Hyperphosphatemia is a serious complication of late-stage chronic kidney disease (CKD). Intestinal inorganic phosphate (Pi) handling plays an important role in Pi homeostasis in CKD. We investigated whether intestinal alkaline phosphatase 3 (Akp3), the enzyme that hydrolyzes dietary Pi compounds, is a target for the treatment of hyperphosphatemia in CKD. METHODS: We investigated Pi homeostasis in Akp3 knockout mice (Akp3-/-). We also studied the progression of renal failure in an Akp3-/- mouse adenine treated renal failure model. Plasma, fecal, and urinary Pi and Ca concentration were measured with commercially available kit, and plasma fibroblast growth factor 23, parathyroid hormone, and 1,25(OH)2D3 concentration were measured with ELISA. Brush border membrane vesicles were prepared from mouse intestine using the Ca2+ precipitation method and used for Pi transport activity and alkaline phosphatase activity. In vivo intestinal Pi absorption was measured with oral 32P administration. RESULTS: Akp3-/- mice exhibited reduced intestinal type II sodium-dependent Pi transporter (Npt2b) protein levels and Na-dependent Pi co-transport activity. In addition, plasma active vitamin D levels were significantly increased in Akp3-/- mice compared with wild-type animals. In the adenine-induced renal failure model, Akp3 gene deletion suppressed hyperphosphatemia. CONCLUSION: The present findings indicate that intestinal Akp3 deletion affects Na+-dependent Pi transport in the small intestine. In the adenine-induced renal failure model, Akp3 is predicted to be a factor contributing to suppression of the plasma Pi concentration.


Assuntos
Fosfatase Alcalina/fisiologia , Homeostase , Fosfatos/metabolismo , Insuficiência Renal/metabolismo , Fosfatase Alcalina/genética , Animais , Transporte Biológico , Modelos Animais de Doenças , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Knockout , Fosfatos/sangue , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/metabolismo
7.
Clin Exp Nephrol ; 22(3): 517-528, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29128884

RESUMO

BACKGROUND: Hyperphosphatemia is common in chronic kidney disease and is associated with morbidity and mortality. The intestinal Na+-dependent phosphate transporter Npt2b is thought to be an important molecular target for the prevention of hyperphosphatemia. The role of Npt2b in the net absorption of inorganic phosphate (Pi), however, is controversial. METHODS: In the present study, we made tamoxifen-inducible Npt2b conditional knockout (CKO) mice to analyze systemic Pi metabolism, including intestinal Pi absorption. RESULTS: Although the Na+-dependent Pi transport in brush-border membrane vesicle uptake levels was significantly decreased in the distal intestine of Npt2b CKO mice compared with control mice, plasma Pi and fecal Pi excretion levels were not significantly different. Data obtained using the intestinal loop technique showed that Pi uptake in Npt2b CKO mice was not affected at a Pi concentration of 4 mM, which is considered the typical luminal Pi concentration after meals in mice. Claudin, which may be involved in paracellular pathways, as well as claudin-2, 12, and 15 protein levels were significantly decreased in the Npt2b CKO mice. Thus, Npt2b deficiency did not affect Pi absorption within the range of Pi concentrations that normally occurs after meals. CONCLUSION: These findings indicate that abnormal Pi metabolism may also be involved in tight junction molecules such as Cldns that are affected by Npt2b deficiency.


Assuntos
Absorção Intestinal , Rim/metabolismo , Fosfatos/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/fisiologia , Animais , Claudinas/metabolismo , Camundongos Knockout , Microvilosidades/metabolismo
8.
Clin Exp Nephrol ; 21(Suppl 1): 21-26, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27900568

RESUMO

The prevention and correction of hyperphosphatemia are major goals of the treatment of chronic kidney disease (CKD)-bone mineral disorders, and thus, Pi balance requires special attention. Pi balance is maintained by intestinal absorption, renal excretion, and bone accretion. The kidney is mainly responsible for the plasma Pi concentration. In CKD, reduced glomerular filtration rate leads to various Pi metabolism abnormalities, and Pi absorption in the small intestine also has an important role in Pi metabolism. Disturbances in Pi metabolism are mediated by a series of complex changes in regulatory hormones originating from the skeleton, intestine, parathyroid gland, and kidney. In this review, we describe the regulation of type II sodium-dependent Pi co-transporters by the kidney and intestine, including the regulation of Pi transport, circadian rhythm, and the vicious circle between salivary Pi secretion and intestinal Pi absorption in animals with and without CKD.


Assuntos
Mucosa Intestinal/metabolismo , Rim/metabolismo , Fosfatos/metabolismo , Animais , Distúrbio Mineral e Ósseo na Doença Renal Crônica/complicações , Distúrbio Mineral e Ósseo na Doença Renal Crônica/metabolismo , Fator de Crescimento de Fibroblastos 23 , Humanos , Minerais/metabolismo , Fosfatos/urina , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/metabolismo
9.
J Bone Miner Metab ; 34(1): 1-10, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26296817

RESUMO

In this review, we focus on the interconnection of inorganic phosphate (Pi) homeostasis in the network of the bone-kidney, parathyroid-kidney, intestine-kidney, and liver-kidney axes. Such a network of organ communication is important for body Pi homeostasis. Normalization of serum Pi levels is a clinical target in patients with chronic kidney disease (CKD). Particularly, disorders of the fibroblast growth factor 23/klotho system are observed in early CKD. Identification of phosphaturic factors from the intestine and liver may enhance our understanding of body Pi homeostasis and Pi metabolism disturbances in CKD patients.


Assuntos
Osso e Ossos/metabolismo , Rim/metabolismo , Fosfatos/metabolismo , Homeostase , Humanos , Mucosa Intestinal/metabolismo , Fígado/metabolismo , Glândulas Paratireoides/metabolismo , Insuficiência Renal Crônica/metabolismo
10.
J Am Soc Nephrol ; 25(4): 761-72, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24262791

RESUMO

Marked hypophosphatemia is common after major hepatic resection, but the pathophysiologic mechanism remains unknown. We used a partial hepatectomy (PH) rat model to investigate the molecular basis of hypophosphatemia. PH rats exhibited hypophosphatemia and hyperphosphaturia. In renal and intestinal brush-border membrane vesicles isolated from PH rats, Na(+)-dependent phosphate (Pi) uptake decreased by 50%-60%. PH rats also exhibited significantly decreased levels of renal and intestinal Na(+)-dependent Pi transporter proteins (NaPi-IIa [NaPi-4], NaPi-IIb, and NaPi-IIc). Parathyroid hormone was elevated at 6 hours after PH. Hyperphosphaturia persisted, however, even after thyroparathyroidectomy in PH rats. Moreover, DNA microarray data revealed elevated levels of nicotinamide phosphoribosyltransferase (Nampt) mRNA in the kidney after PH, and Nampt protein levels and total NAD concentration increased significantly in the proximal tubules. PH rats also exhibited markedly increased levels of the Nampt substrate, urinary nicotinamide (NAM), and NAM catabolites. In vitro analyses using opossum kidney cells revealed that NAM alone did not affect endogenous NaPi-4 levels. However, in cells overexpressing Nampt, the addition of NAM led to a marked decrease in cell surface expression of NaPi-4 that was blocked by treatment with FK866, a specific Nampt inhibitor. Furthermore, FK866-treated mice showed elevated renal Pi reabsorption and hypophosphaturia. These findings indicate that hepatectomy-induced hypophosphatemia is due to abnormal NAM metabolism, including Nampt activation in renal proximal tubular cells.


Assuntos
Hepatectomia/efeitos adversos , Hipofosfatemia/etiologia , Rim/metabolismo , Acrilamidas/farmacologia , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NAD/metabolismo , Niacinamida/metabolismo , Nicotinamida Fosforribosiltransferase/fisiologia , Paratireoidectomia , Piperidinas/farmacologia , Ratos , Ratos Wistar , Proteínas Cotransportadoras de Sódio-Fosfato/fisiologia , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/fisiologia
11.
Clin Calcium ; 25(7): 1043-7, 2015 Jul.
Artigo em Japonês | MEDLINE | ID: mdl-26119318

RESUMO

Osteocytes orchestrate bone resorption and bone formation by controlling osteoclast and osteoblast activity. On the other hand, osteocytes secret FGF23 (fibroblast growth factor 23), FGF23 acts on the kidney to control phosphate homeostasis. Sclerostin is also released from osteocytes and it regulated osteoblast activity through Wnt/ß-catenin pathway. Therefore, an antibody that targets sclerostin is currently in phase- III clinical trials for the treatment of osteoporosis and it is expected as new therapeutics.


Assuntos
Proteínas Morfogenéticas Ósseas/imunologia , Proteínas Morfogenéticas Ósseas/fisiologia , Osso e Ossos/metabolismo , Marcadores Genéticos/imunologia , Marcadores Genéticos/fisiologia , Osteócitos/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Anticorpos Monoclonais/uso terapêutico , Proteínas Morfogenéticas Ósseas/metabolismo , Reabsorção Óssea , Cálcio/metabolismo , Ensaios Clínicos Fase III como Assunto , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/metabolismo , Homeostase , Humanos , Rim/metabolismo , Camundongos , Terapia de Alvo Molecular , Osteoblastos/fisiologia , Osteoclastos/fisiologia , Osteócitos/metabolismo , Osteogênese/genética , Osteoporose/tratamento farmacológico , Fosfatos/metabolismo , Via de Sinalização Wnt/fisiologia , beta Catenina/fisiologia
12.
Clin Calcium ; 24(2): 249-55, 2014 Feb.
Artigo em Japonês | MEDLINE | ID: mdl-24473358

RESUMO

Phosphate (Pi), one of most abundant anions in living organisms, plays a crucial role in biomineralization. An adequate plasma Pi concentration is required to maintain the calcium × phosphate ion product within a range sufficient for physiological bone mineralization, but an increase in the calcium × phosphate product in extracellular fluids above a certain threshold can predispose to extraskeletal calcification. Membrane transport systems for Pi transport are key elements in maintaining homeostasis of Pi in organisms. Members of two families of solute carrier (SLC) proteins (SLC20 and SLC34) act as Na⁺ -dependent, secondary-active cotransporters to transport Pi across cell membranes in mammals. This review summarizes the role of SLC20 and SCL34 proteins on biomineralization.


Assuntos
Calcificação Fisiológica , Transporte de Íons/fisiologia , Fosfatos/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato/metabolismo , Sódio/metabolismo , Animais , Homeostase/fisiologia , Humanos , Transporte de Íons/genética , Proteínas Cotransportadoras de Sódio-Fosfato/genética
13.
Am J Physiol Cell Physiol ; 302(9): C1316-30, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22159077

RESUMO

Mutations in the apically located Na(+)-dependent phosphate (NaPi) cotransporter, SLC34A3 (NaPi-IIc), are a cause of hereditary hypophosphatemic rickets with hypercalciuria (HHRH). We have characterized the impact of several HHRH mutations on the processing and stability of human NaPi-IIc. Mutations S138F, G196R, R468W, R564C, and c.228delC in human NaPi-IIc significantly decreased the levels of NaPi cotransport activities in Xenopus oocytes. In S138F and R564C mutant proteins, this reduction is a result of a decrease in the V(max) for P(i), but not the K(m). G196R, R468W, and c.228delC mutants were not localized to oocyte membranes. In opossum kidney (OK) cells, cell surface labeling, microscopic confocal imaging, and pulse-chase experiments showed that G196R and R468W mutations resulted in an absence of cell surface expression owing to endoplasmic reticulum (ER) retention. G196R and R468W mutants could be partially stabilized by low temperature. In blue native-polyacrylamide gel electrophoresis analysis, G196R and R468W mutants were either denatured or present in an aggregation complex. In contrast, S138F and R564C mutants were trafficked to the cell surface, but more rapidly degraded than WT protein. The c.228delC mutant did not affect endogenous NaPi uptake in OK cells. Thus, G196R and R468W mutations cause ER retention, while S138F and R564C mutations stimulate degradation of human NaPi-IIc in renal epithelial cells. Together, these data suggest that the NaPi-IIc mutants in HHRH show defective processing and stability.


Assuntos
Raquitismo Hipofosfatêmico Familiar/genética , Raquitismo Hipofosfatêmico Familiar/metabolismo , Hipercalciúria/genética , Hipercalciúria/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIc/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIc/metabolismo , Animais , Western Blotting , Humanos , Imunoprecipitação , Microscopia Confocal , Mutagênese Sítio-Dirigida , Mutação , Reação em Cadeia da Polimerase , Estabilidade Proteica , Transfecção , Xenopus laevis
14.
Clin Calcium ; 22(10): 1537-41, 2012 Oct.
Artigo em Japonês | MEDLINE | ID: mdl-23023634

RESUMO

Small intestine plays an important role in the sensing of various nutrients. There is information that would imply the existence of a dietary phosphate sensing mechanism within the intestine. Recent studies suggest that intestinal factors may involve in the alteration of renal phosphate transport. The elucidation of the phosphate sensing mechanism is expected to provide molecular basis for the prevention of the hyperphosphatemia in chronic kidney disease patients.


Assuntos
Homeostase/fisiologia , Intestino Delgado/metabolismo , Fosfatos/metabolismo , Insuficiência Renal Crônica/metabolismo , Transdução de Sinais/fisiologia , Animais , Humanos , Hiperfosfatemia/prevenção & controle
15.
Clin Calcium ; 22(10): 1469-76, 2012 Oct.
Artigo em Japonês | MEDLINE | ID: mdl-23023625

RESUMO

Inorganic phosphate (Pi) is an essential nutrient for several biological functions, including intracellular signal transduction, the production and function of cell membranes, and energy exchange. To achieve these functions, a transport system is required to transfer Pi across hydrophobic cell membranes. Pi (re) absorption in the small intestine and renal proximal tubules is important for Pi homeostasis. Three types of NaPi transporters (types I - III ) have been identified : solute carrier series SLC17A1 (NPT1/NaPi- I /OATv1) , SLC34 (NaPi- II a, NaPi- II b, NaPi- II c) , and SLC20 (PiT1, PiT2) , respectively. In this review, we discuss the role of NaPi transporters in Pi homeostasis.


Assuntos
Homeostase , Mucosa Intestinal/metabolismo , Rim/metabolismo , Fosfatos/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato/metabolismo , Sódio/metabolismo , Animais , Humanos
16.
Clin Calcium ; 22(10): 1583-91, 2012 Oct.
Artigo em Japonês | MEDLINE | ID: mdl-23023640

RESUMO

Hyperphosphatemia is a common disorder in patients with chronic kidney disease (CKD) , and may result in hyperparathyroidism and renal osteodystrophy. Hyperphosphatemia also may contribute to deterioration vascular calcification and increase mortality. Hence, correction and prevention of hyperphosphatemia is a main component of the management of CKD. This goal is usually approached both by administering phosphorus binders and by restricting dietary phosphorus (P) intake. Dietary intake of phosphorus (P) is derived largely from foods with high protein content or food additives and is an important determinant of P balance in patient with CKD. Food additives (PO4) can dramatically increase the amount of P consumed in the daily diet, especially because P is more readily absorbed in its inorganic form. In addition, information about the P content and type in prepared foods is often unavailable or misleading. Therefore, during dietary counseling of patients with CKD, we recommended that they consider both the absolute dietary P content and the P-to-protein ratio of foods and meals including food additives.


Assuntos
Aditivos Alimentares/efeitos adversos , Hiperfosfatemia/etiologia , Fósforo na Dieta/efeitos adversos , Fósforo/efeitos adversos , Aditivos Alimentares/normas , Humanos , Hiperfosfatemia/complicações , Fósforo/administração & dosagem , Fósforo na Dieta/administração & dosagem , Diálise Renal , Insuficiência Renal Crônica/etiologia
17.
Am J Physiol Renal Physiol ; 301(5): F1105-13, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21816756

RESUMO

An inorganic phosphate (P(i))-restricted diet is important for patients with chronic kidney disease and patients on hemodialysis. Phosphate binders are essential for preventing hyperphosphatemia and ectopic calcification. The sodium-dependent P(i) (Na/P(i)) transport system is involved in intestinal P(i) absorption and is regulated by several factors. The type II sodium-dependent P(i) transporter Npt2b is expressed in the brush-border membrane in intestinal epithelial cells and transports P(i). In the present study, we analyzed the phenotype of Npt2b(-/-) and hetero(+/-) mice. Npt2b(-/-) mice died in utero soon after implantation, indicating that Npt2b is essential for early embryonic development. At 4 wk of age, Npt2b(+/-) mice showed hypophosphatemia and low urinary P(i) excretion. Plasma fibroblast growth factor 23 levels were significantly decreased and 1,25(OH)(2)D(3) levels were significantly increased in Npt2b(+/-) mice compared with Npt2b(+/+) mice. Npt2b mRNA levels were reduced to 50% that in Npt2b(+/+) mice. In contrast, renal Npt2a and Npt2c transporter protein levels were significantly increased in Npt2b(+/-) mice. At 20 wk of age, Npt2b(+/-) mice showed hypophosphaturia and reduced Na/P(i) cotransport activity in the distal intestine. Npt2b(+/+) mice with adenine-induced renal failure had hyperphosphatemia and high plasma creatinine levels. Npt2b(+/-) mice treated with adenine had significantly reduced plasma P(i) levels compared with Npt2b(+/+) mice. Intestinal Npt2b protein and Na(+)/P(i) transport activity levels were significantly lower in Npt2b(+/-) mice than in the Npt2b(+/+) mice. The findings of the present studies suggest that Npt2b is an important target for the prevention of hyperphosphatemia.


Assuntos
Homeostase/fisiologia , Fosfatos/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/fisiologia , Adenina , Animais , Western Blotting , Peso Corporal/fisiologia , Cromossomos Artificiais Bacterianos/genética , DNA/genética , Dieta , Feminino , Vetores Genéticos , Genótipo , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microvilosidades/metabolismo , Fosfatos/sangue , Reação em Cadeia da Polimerase , Gravidez , Insuficiência Renal/induzido quimicamente , Insuficiência Renal/metabolismo , Sódio/metabolismo
18.
Am J Physiol Renal Physiol ; 299(1): F243-54, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20410212

RESUMO

The type IIc Na(+)-dependent phosphate cotransporter (NaPi-IIc) is specifically targeted to, and expressed on, the apical membrane of renal proximal tubular cells and mediates phosphate transport. In the present study, we investigated the signals that determine apical expression of NaPi-IIc with a focus on the role of the N- and the C-terminal tails of mouse NaPi-IIc in renal epithelial cells [opossum kidney (OK) and Madin-Darby canine kidney cells]. Wild-type NaPi-IIc, the cotransporter NaPi-IIa, as well as several IIa-IIc chimeras and deletion mutants, were fused to enhanced green fluorescent protein (EGFP), and their cellular localization was analyzed in polarized renal epithelial cells by confocal microscopy and by cell-surface biotinylation. Fluorescent EGFP-fused NaPi-IIc transporter proteins are correctly expressed in the apical membrane of OK cells. The apical expression of N-terminal deletion mutants (deletion of N-terminal 25, 50, or 69 amino acids) was not affected by truncation. In contrast, C-terminal deletion mutants (deletion of C-terminal 45, 50, or 62 amino acids) did not have correct apical expression. A more detailed mutational analysis indicated that a domain (amino acids WLHSL) in the cytoplasmic C terminus is required for apical expression of NaPi-IIc in renal epithelial cells. We conclude that targeting of NaPi-IIc to the apical cell surface is regulated by a unique amino acid motif in the cytoplasmic C-terminal domain.


Assuntos
Polaridade Celular , Células Epiteliais/metabolismo , Rim/metabolismo , Sinais Direcionadores de Proteínas , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIc/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Biotinilação , Células CACO-2 , Cães , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Rim/citologia , Células LLC-PK1 , Camundongos , Microscopia Confocal , Dados de Sequência Molecular , Mutação , Gambás , Estrutura Terciária de Proteína , Transporte Proteico , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIc/genética , Suínos , Transfecção
19.
Am J Physiol Renal Physiol ; 298(6): F1341-50, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20357029

RESUMO

In the present study, we evaluated the roles of type II and type III sodium-dependent P(i) cotransporters in fibroblast growth factor 23 (FGF23) activity by administering a vector encoding FGF23 with the R179Q mutation (FGF23M) to wild-type (WT) mice, Npt2a knockout (KO) mice, Npt2c KO mice, and Npt2a(-/-)Npt2c(-/-) mice (DKO mice). In Npt2a KO mice, FGF23M induced severe hypophosphatemia and markedly decreased the levels of Npt2c, type III Na-dependent P(i) transporter (PiT2) protein, and renal Na/P(i) transport activity. In contrast, in Npt2c KO mice, FGF23M decreased plasma phosphate levels comparable to those in FGF23M-injected WT mice. In DKO mice with severe hypophosphatemia, FGF23M administration did not induce an additional increase in urinary phosphate excretion. FGF23 administration significantly decreased intestinal Npt2b protein levels in WT mice but had no effect in Npt2a, Npt2c, and DKO mice, despite marked suppression of plasma 1,25(OH)(2)D(3) levels in all the mutant mice. The main findings were as follow: 1) FGF23-dependent phosphaturic activity in Npt2a KO mice is dependent on renal Npt2c and PiT-2 protein; 2) in DKO mice, renal P(i) reabsorption is not further decreased by FGF23M, but renal vitamin D synthesis is suppressed; and 3) downregulation of intestinal Npt2b may be mediated by a factor(s) other than 1,25(OH)(2)D(3). These findings suggest that Npt2a, Npt2c, and PiT-2 are necessary for the phosphaturic activity of FGF23. Thus complementary regulation of Npt2 family proteins may be involved in systemic P(i) homeostasis.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Hipofosfatemia Familiar/etiologia , Hipofosfatemia/etiologia , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/deficiência , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIc/deficiência , Animais , Calcitriol/sangue , Cálcio/sangue , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/sangue , Fatores de Crescimento de Fibroblastos/genética , Técnicas de Transferência de Genes , Humanos , Hipofosfatemia/genética , Hipofosfatemia/metabolismo , Hipofosfatemia Familiar/genética , Hipofosfatemia Familiar/metabolismo , Injeções Intravenosas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Fosfatos/sangue , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIc/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA