Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros

Medicinas Complementares
Métodos Terapêuticos e Terapias MTCI
Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Biochem Soc Trans ; 50(1): 439-446, 2022 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-34994388

RESUMO

Hyperphosphatemia results from an imbalance in phosphate (Pi) homeostasis. In patients with and without reduced kidney function, hyperphosphatemia is associated with cardiovascular complications. The current mainstays in the management of hyperphosphatemia are oral Pi binder and dietary Pi restriction. Although these options are employed in patients with chronic kidney disease (CKD), they seem inadequate to correct elevated plasma Pi levels. In addition, a paradoxical increase in expression of intestinal Pi transporter and uptake may occur. Recently, studies in rodents targeting the renal Na+/Pi cotransporter 2a (Npt2a), responsible for ∼70% of Pi reabsorption, have been proposed as a potential treatment option. Two compounds (PF-06869206 and BAY-767) have been developed which are selective for Npt2a. These Npt2a inhibitors significantly increased urinary Pi excretion consequently lowering plasma Pi and PTH levels. Additionally, increases in urinary excretions of Na+, Cl- and Ca2+ have been observed. Some of these results are also seen in models of reduced kidney function. Responses of FGF23, a phosphaturic hormone that has been linked to the development of left ventricular hypertrophy in CKD, are ambiguous. In this review, we discuss the recent advances on the role of Npt2a inhibition on Pi homeostasis as well as other pleiotropic effects observed with Npt2a inhibition.


Assuntos
Hiperfosfatemia , Insuficiência Renal Crônica , Animais , Feminino , Humanos , Hiperfosfatemia/tratamento farmacológico , Masculino , Camundongos , Camundongos Knockout , Hormônio Paratireóideo/metabolismo , Fosfatos/metabolismo , Insuficiência Renal Crônica/tratamento farmacológico , Insuficiência Renal Crônica/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/metabolismo
2.
Ann Anat ; 221: 27-37, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30240906

RESUMO

In a rat model of the andropause we aimed to examine the influence of daidzein, soy isoflavone, on the structure and function of parathyroid glands (PTG) and the expression levels of some of the crucial regulators of Ca2+ and Pi homeostasis in the kidney, and to compare these effects with the effects of estradiol, serving as a positive control. Middle-aged (16-month-old) male Wistar rats were divided into the following groups: sham-operated (SO), orchidectomized (Orx), orchidectomized and estradiol-treated (Orx+E; 0.625mg/kg b.w./day, s.c.) as well as orchidectomized and daidzein-treated (Orx+D; 30mg/kg b.w./day, s.c.) group. Every treated group had a corresponding control group. PTH serum concentration was decreased in Orx+E and Orx+D groups by 10% and 21% (p<0.05) respectively, in comparison with the Orx. PTG volume was decreased in Orx+E group by 16% (p<0.05), when compared to the Orx. In Orx+E group expression of NaPi 2a was lower (p<0.05), while NaPi 2a abundance in Orx+D animals was increased (p<0.05), when compared to Orx. Expression of PTH1R was increased (p<0.05) in Orx+E group, while in Orx+D animals the same parameter was decreased (p<0.05), in comparison with Orx. Klotho expression was elevated (p<0.05) in Orx+D rats, in regard to Orx. Orx+D induced reduction in Ca2+/creatinine and Pi/creatinine ratio in urine by 32% and 16% (p<0.05) respectively, in comparison with Orx. In conclusion, presented results indicate the more coherent beneficial effects of daidzein compared to estradiol, on disturbed Ca2+ and Pi homeostasis, and presumably on bone health, in the aging male rats.


Assuntos
Andropausa , Modelos Animais de Doenças , Glucuronidase/efeitos dos fármacos , Isoflavonas/farmacologia , Fitoestrógenos/farmacologia , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/efeitos dos fármacos , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Glucuronidase/genética , Glucuronidase/metabolismo , Proteínas Klotho , Masculino , Orquiectomia , Ratos , Ratos Wistar , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/metabolismo , Regulação para Cima
3.
Biomed Pharmacother ; 106: 54-60, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29957466

RESUMO

Chronic kidney disease-mineral and bone disorder (CKD-MBD) play a critical role in the pathogenesis of cardiovascular complications in patients with chronic kidney disease (CKD). Zuogui pill as a traditional Chinese herbal drug has been used for nourish kidney essence improve bone malnutrition of renal bone disease by regulating the metabolism of calcium and phosphorus and participating in osteoblast metabolism. In the present study, 5/6 nephrectomy rat model was used to reveal the mechanism of zuogui pill in treatment of CKD-MBD. Compared with sham rats, the levels of serum phosphorus, PTH, iPTH and creatinine were significantly decreased, while the serum calcium level was significantly increased, and the Cbfa1 protein level was significantly decreased and FGF23 protein level was significantly increased by Zuogui pill treatment. Compared with model rats, the BMD of rat was significantly increased by Zuogui pill treatment. Histological analysis revealed that the kidney injury of rats with CKD was significantly reduced by zuogui pill treatment. Compared with model rats, the CYP27B1 mRNA level was significantly increased, and the PTH mRNA level and NaPiIIa protein level were significantly decreased in the kidney by zuogui pill treatment. We inferred that zuogui pill exhibited potential therapeutic effects on CKD-MBD in the rats by regulating bone metabolism and nourish kidney.


Assuntos
Remodelação Óssea/efeitos dos fármacos , Osso e Ossos/efeitos dos fármacos , Distúrbio Mineral e Ósseo na Doença Renal Crônica/tratamento farmacológico , Medicamentos de Ervas Chinesas/farmacologia , Rim/efeitos dos fármacos , 25-Hidroxivitamina D3 1-alfa-Hidroxilase/genética , 25-Hidroxivitamina D3 1-alfa-Hidroxilase/metabolismo , Animais , Densidade Óssea/efeitos dos fármacos , Osso e Ossos/metabolismo , Osso e Ossos/fisiopatologia , Cálcio/sangue , Distúrbio Mineral e Ósseo na Doença Renal Crônica/sangue , Distúrbio Mineral e Ósseo na Doença Renal Crônica/patologia , Distúrbio Mineral e Ósseo na Doença Renal Crônica/fisiopatologia , Subunidade alfa 1 de Fator de Ligação ao Core/sangue , Creatinina/sangue , Modelos Animais de Doenças , Fatores de Crescimento de Fibroblastos/sangue , Rim/metabolismo , Rim/patologia , Rim/fisiopatologia , Hormônio Paratireóideo/sangue , Hormônio Paratireóideo/genética , Fósforo/sangue , Ratos Wistar , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/metabolismo
4.
Endocrinology ; 158(2): 252-263, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-27929669

RESUMO

Loss of fibroblast growth factor-23 (FGF23) causes hyperphosphatemia, extraskeletal calcifications, and early mortality; excess FGF23 causes hypophosphatemia with rickets or osteomalacia. However, FGF23 may not be important during fetal development. FGF23 deficiency (Fgf23 null) and FGF23 excess (Phex null) did not alter fetal phosphorus or skeletal parameters. In this study, we further tested our hypothesis that FGF23 is not essential for fetal phosphorus regulation but becomes important after birth. Although coreceptor Klotho null adults have extremely high FGF23 concentrations, intact FGF23 was normal in Klotho null fetuses, as were fetal phosphorus and skeletal parameters and placental and renal expression of FGF23 target genes. Pth/Fgf23 double mutants had the same elevation in serum phosphorus as Pth null fetuses, as compared with normal serum phosphorus in Fgf23 nulls. We examined the postnatal time courses of Fgf23 null, Klotho null, and Phex null mice. Fgf23 nulls and Klotho nulls were normal at birth, but developed hyperphosphatemia, increased renal expression of NaPi2a and NaPi2c, and reduced renal phosphorus excretion between 5 and 7 days after birth. Parathyroid hormone remained normal. In contrast, excess FGF23 exerted effects in Phex null males within 12 hours after birth, with the development of hypophosphatemia, reduced renal expression of NaPi2a and NaPi2c, and increased renal phosphorus excretion. In conclusion, although FGF23 is present in the fetal circulation at levels that may equal adult values, and there is robust expression of FGF23 target genes in placenta and fetal kidneys, FGF23 itself is not an important regulator of fetal phosphorous metabolism.


Assuntos
Feto/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Fósforo/sangue , Animais , Animais Recém-Nascidos , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/genética , Glucuronidase/genética , Glucuronidase/metabolismo , Rim/metabolismo , Proteínas Klotho , Masculino , Camundongos Endogâmicos C57BL , Endopeptidase Neutra Reguladora de Fosfato PHEX/genética , Hormônio Paratireóideo/sangue , Fenótipo , Gravidez , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/metabolismo
5.
Am J Physiol Renal Physiol ; 312(1): F77-F83, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27784695

RESUMO

Mutations in the renal sodium-dependent phosphate cotransporters NPT2a and NPT2c have been reported in patients with renal stone disease and nephrocalcinosis. Oral phosphate supplementation is currently thought to reduce risk by reversing the hypercalciuria, but the exact mechanism remains unclear and the relative contribution of modifiers of mineralization such as osteopontin (Opn) to the formation of renal mineral deposits in renal phosphate wasting disorders has not been studied. We observed a marked decrease of renal gene expression and urinary excretion of Opn in Npt2a-/- mice, a mouse model of these disorders, at baseline. Following supplementation with phosphate Opn gene expression was restored to wild-type levels in Npt2a-/- mice; however, urine excretion of the protein remained low. To further investigate the role of Opn, we used a double-knockout strategy, which provides evidence that loss of Opn worsens the nephrocalcinosis and nephrolithiasis observed in these mice on a high-phosphate diet. These studies suggest that impaired Opn gene expression and urinary excretion in Npt2a-/- mice may be an additional risk factor for nephrolithiasis, and normalizing urine Opn levels may improve the therapy of phosphaturic disorders.


Assuntos
Raquitismo Hipofosfatêmico Familiar/metabolismo , Hipercalciúria/metabolismo , Rim/metabolismo , Nefrocalcinose/metabolismo , Osteopontina/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/genética , Animais , Feminino , Fatores de Crescimento de Fibroblastos/genética , Hipofosfatemia/genética , Masculino , Camundongos Knockout , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIc/genética
6.
J Bone Miner Res ; 31(5): 929-39, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26751835

RESUMO

X-linked hypophosphatemia (XLH) is characterized by impaired renal tubular reabsorption of phosphate owing to increased circulating FGF23 levels, resulting in rickets in growing children and impaired bone mineralization. Increased FGF23 decreases renal brush border membrane sodium-dependent phosphate transporter IIa (Npt2a) causing renal phosphate wasting, impairs 1-α hydroxylation of 25-hydroxyvitamin D, and induces the vitamin D 24-hydroxylase, leading to inappropriately low circulating levels of 1,25-dihydroxyvitamin D (1,25D). The goal of therapy is prevention of rickets and improvement of growth in children by phosphate and 1,25D supplementation. However, this therapy is often complicated by hypercalcemia and nephrocalcinosis and does not always prevent hyperparathyroidism. To determine if 1,25D or blocking FGF23 action can improve the skeletal phenotype without phosphate supplementation, mice with XLH (Hyp) were treated with daily 1,25D repletion, FGF23 antibodies (FGF23Ab), or biweekly high-dose 1,25D from d2 to d75 without supplemental phosphate. All treatments maintained normocalcemia, increased serum phosphate, and normalized parathyroid hormone levels. They also prevented the loss of Npt2a, α-Klotho, and pERK1/2 immunoreactivity observed in the kidneys of untreated Hyp mice. Daily treatment with 1,25D decreased urine phosphate losses despite a marked increase in bone FGF23 mRNA and in circulating FGF23 levels. Daily 1,25D was more effective than other treatments in normalizing the growth plate and metaphyseal organization. In addition to being the only therapy that normalized lumbar vertebral height and body weight, daily 1,25D therapy normalized bone geometry and was more effective than FGF23Ab in improving trabecular bone structure. Daily 1,25D and FGF23Ab improved cortical microarchitecture and whole-bone biomechanical properties more so than biweekly 1,25D. Thus, monotherapy with 1,25D improves growth, skeletal microarchitecture, and bone strength in the absence of phosphate supplementation despite enhancing FGF23 expression, demonstrating that 1,25D has direct beneficial effects on the skeleton in XLH, independent of its role in phosphate homeostasis. © 2016 American Society for Bone and Mineral Research.


Assuntos
Densidade Óssea/efeitos dos fármacos , Calcitriol/farmacologia , Raquitismo Hipofosfatêmico Familiar , Fatores de Crescimento de Fibroblastos/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Esqueleto , Animais , Modelos Animais de Doenças , Raquitismo Hipofosfatêmico Familiar/tratamento farmacológico , Raquitismo Hipofosfatêmico Familiar/metabolismo , Raquitismo Hipofosfatêmico Familiar/patologia , Fator de Crescimento de Fibroblastos 23 , Humanos , Camundongos , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Esqueleto/metabolismo , Esqueleto/patologia , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/metabolismo
7.
J Comp Physiol B ; 184(1): 137-47, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24129944

RESUMO

In our former studies low crude protein (LCP) intake influenced N homeostasis and electrolyte handling in goats. We hypothesised that due to rumino-hepatic nitrogen (N) recycling adaptation of N homeostasis and adjustment of electrolyte handling to LCP intake differs between goats and monogastric animals. Therefore, an experiment similar to that with goats was conducted with rats. Two feeding groups received a diet either containing 20 or 8 % crude protein (as fed basis) for 5 weeks and intake and excretion of N, calcium (Ca) and phosphorus (P) were determined. To detect systemic and endocrine adaptation to LCP intake plasma concentrations of urea, Ca, phosphate (Pi), insulin-like growth factor 1 (IGF-1), 1,25-dihydroxyvitamin D3 (calcitriol), parathyroid hormone (PTH) and cross-linked telopeptide of type I collagen (CTX) were measured. Adjustment of renal electrolyte transport was assessed by detecting protein expression of key proteins of renal Pi transport. All data were compared with the data of the goat experiment. LCP intake decreased plasma urea concentration stronger in goats than in rats. In both species urinary N excretion declined, but faecal N excretion decreased in goats only. Furthermore, in goats urinary Ca excretion decreased, but in rats urinary Ca concentration increased. Decreased plasma IGF-1 and calcitriol concentrations were found in goats only. Thus, renal Ca excretion appears to be a common target in adaptation of electrolyte homeostasis in both species, but is regulated differently.


Assuntos
Cálcio/urina , Proteínas Alimentares/farmacologia , Cabras/metabolismo , Nitrogênio/metabolismo , Fósforo/urina , Ratos/metabolismo , Animais , Calcitriol/sangue , Cálcio/sangue , Eletrólitos/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fezes/química , Homeostase , Fator de Crescimento Insulin-Like I/metabolismo , Rim/efeitos dos fármacos , Rim/metabolismo , Masculino , Hormônio Paratireóideo/sangue , Ratos Wistar , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/metabolismo , Especificidade da Espécie
8.
Eur J Pharmacol ; 721(1-3): 332-40, 2013 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-24056120

RESUMO

Hyperphosphatemia is associated with severe decline of renal function in chronic kidney disease and elevates cardiovascular mortality. Type II sodium dependent phosphate transporter 2A (Npt2A) plays a major role in renal phosphate reabsorption and could be explored as a target for anti-hyperphosphatemia therapy. Human Npt2A transporter activity was examined upon transfection into CHO, MDCK, HEK293, Flp-In-CHO and Flp-In-HEK293 cells. Only kidney-derived cells expressed functional Npt2A. HEK293 and Flp-In-HEK293 cell lines stably transfected with hNpt2A could be selected, but these cells were inactive in phosphate transport. This suggests that high-level, constitutive Npt2A expression has deleterious effects on the cell. By using the conditional promoter in the Flp-In-Trex vector, functional expression of Npt2A was achieved by doxycycline induction in HEK293 cells. The EGFP tagged and non-tagged, inducible stable hNpt2A-HEK293 cell lines afforded development of a robust phosphate uptake assay mediated by hNpt2A, which can be used to screen hNpt2A inhibitors and inducers of hNpt2A expression. Using this assay, the small molecule LC-1 was identified as a potent inhibitor of hNpt2A, suggesting that it is feasible to develop potent specific hNpt2A inhibitors to control phosphate overloading for hyperphosphatemia therapy.


Assuntos
Bioensaio/métodos , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/metabolismo , Transporte Biológico/efeitos dos fármacos , Relação Dose-Resposta a Droga , Doxiciclina/farmacologia , Avaliação Pré-Clínica de Medicamentos , Células HEK293 , Humanos , Fosfatos/metabolismo , Sesquiterpenos/farmacologia , Sódio/farmacologia , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/antagonistas & inibidores , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/genética , Fatores de Tempo , Transfecção
9.
J Physiol Pharmacol ; 64(3): 361-8, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23959733

RESUMO

This study aimed to examine the effects of genistein on the structural and functional changes in parathyroid glands (PTG) and sodium phosphate cotransporter 2a (NaPi 2a) in orchidectomized rats. Sixteen-month-old Wistar rats were divided into sham-operated (SO), orchidectomized (Orx) and genistein-treated orchidectomized (Orx+G) groups. Genistein (30 mg/kg/day) was administered subcutaneously for 3 weeks, while the controls received vehicle alone. PTG was analyzed histomorphometrically, while the expressions of NaPi 2a mRNA/protein levels from kidneys were determined by real time PCR and Western blots. Serum and urine parameters were determined biochemically. The PTG volume in Orx rats was increased by 30% (p<0.05), compared to the SO group. Orx+G treatment increased the PTG volume by 35% and 75% (p<0.05) respectively, comparing to Orx and SO animals. Orchidectomy led to increment of serum PTH by 27% (p<0.05) compared to the SO group, Orx+G decreased it by 18% (p<0.05) comparing to Orx animals. NaPi 2a expression in Orx animals was reduced in regards to its abundance in SO animals, although it was increased in Orx+G group compared to the Orx. Phosphorus urine content of Orx animals was raised by 12% (p<0.05) compared to that for the SO group, while Orx+G induced a 17% reduction (p<0.05) in regards to Orx animals. Our study shows that Orx increases PTG volume and serum PTH level, while protein expression of NaPi 2a is reduced. Application of genistein attenuates the orchidectomy-induced changes in serum PTH level, stimulates the expression of NaPi 2a and reduces urinary Pi excretion, implying potential beneficial effects on andropausal symptoms.


Assuntos
Andropausa , Genisteína/uso terapêutico , Rim/efeitos dos fármacos , Glândulas Paratireoides/efeitos dos fármacos , Fitoestrógenos/uso terapêutico , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/metabolismo , Desequilíbrio Hidroeletrolítico/prevenção & controle , Animais , Cálcio/sangue , Cálcio/urina , Regulação da Expressão Gênica/efeitos dos fármacos , Genisteína/administração & dosagem , Hipocalcemia/etiologia , Hipocalcemia/prevenção & controle , Hipofosfatemia/etiologia , Hipofosfatemia/prevenção & controle , Injeções Subcutâneas , Rim/crescimento & desenvolvimento , Rim/metabolismo , Rim/ultraestrutura , Masculino , Orquiectomia/efeitos adversos , Tamanho do Órgão/efeitos dos fármacos , Glândulas Paratireoides/crescimento & desenvolvimento , Glândulas Paratireoides/metabolismo , Glândulas Paratireoides/ultraestrutura , Hormônio Paratireóideo/sangue , Fósforo/sangue , Fósforo/urina , Fitoestrógenos/administração & dosagem , Ratos , Ratos Wistar , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/biossíntese , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/genética , Desequilíbrio Hidroeletrolítico/etiologia , Desequilíbrio Hidroeletrolítico/metabolismo , Desequilíbrio Hidroeletrolítico/fisiopatologia
10.
J Biol Chem ; 286(2): 1618-26, 2011 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-21047792

RESUMO

The parathyroid hormone (PTH)/PTH-related peptide (PTHrP) receptor (PTHR1) in cells of the renal proximal tubule mediates the reduction in membrane expression of the sodium-dependent P(i) co-transporters, NPT2a and NPT2c, and thus suppresses the re-uptake of P(i) from the filtrate. In most cell types, the liganded PTHR1 activates Gα(S)/adenylyl cyclase/cAMP/PKA (cAMP/PKA) and Gα(q/11)/phospholipase C/phosphatidylinositol 1,4,5-trisphosphate (IP(3))/Ca(2+)/PKC (IP(3)/PKC) signaling pathways, but the relative roles of each pathway in mediating renal regulation P(i) transport remain uncertain. We therefore explored the signaling mechanisms involved in PTH-dependent regulation of NPT2a function using potent, long-acting PTH analogs, M-PTH(1-28) (where M = Ala(1,12), Aib(3), Gln(10), Har(11), Trp(14), and Arg(19)) and its position 1-modified variant, Trp(1)-M-PTH(1-28), designed to be phospholipase C-deficient. In cell-based assays, both M-PTH(1-28) and Trp(1)-M-PTH(1-28) exhibited potent and prolonged cAMP responses, whereas only M-PTH(1-28) was effective in inducing IP(3) and intracellular calcium responses. In opossum kidney cells, a clonal cell line in which the PTHR1 and NPT2a are endogenously expressed, M-PTH(1-28) and Trp(1)-M-PTH(1-28) each induced reductions in (32)P uptake, and these responses persisted for more than 24 h after ligand wash-out, whereas that of PTH(1-34) was terminated by 4 h. When injected into wild-type mice, both M-modified PTH analogs induced prolonged reductions in blood P(i) levels and commensurate reductions in NPT2a expression in the renal brush border membrane. Our findings suggest that the acute down-regulation of NPT2a expression by PTH ligands involves mainly the cAMP/PKA signaling pathway and are thus consistent with the elevated blood P(i) levels seen in pseudohypoparathyroid patients, in whom Gα(s)-mediated signaling in renal proximal tubule cells is defective.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Hormônio Paratireóideo/metabolismo , Pseudo-Hipoparatireoidismo/metabolismo , Transdução de Sinais/fisiologia , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/metabolismo , Animais , Células COS , Bovinos , Chlorocebus aethiops , Regulação para Baixo/fisiologia , Humanos , Técnicas In Vitro , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Gambás , Osteoblastos/citologia , Osteoblastos/metabolismo , Hormônio Paratireóideo/análogos & derivados , Hormônio Paratireóideo/genética , Fósforo/sangue , Ratos , Sódio/metabolismo
11.
Am J Physiol Renal Physiol ; 297(2): F282-91, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19515808

RESUMO

Fibroblast growth factor-23 (FGF23) is a phosphaturic hormone that contributes to several hypophosphatemic disorders by reducing the expression of the type II sodium-phosphate cotransporters (NaPi-2a and NaPi-2c) in the kidney proximal tubule and by reducing serum 1,25-dihydroxyvitamin D(3) [1,25(OH)(2)D(3)] levels. The FGF receptor(s) mediating the hypophosphatemic action of FGF23 in vivo have remained elusive. In this study, we show that proximal tubules express FGFR1, -3, and -4 but not FGFR2 mRNA. To determine which of these three FGFRs mediates FGF23's hypophosphatemic actions, we characterized phosphate homeostasis in FGFR3(-/-) and FGFR4(-/-) null mice, and in conditional FGFR1(-/-) mice, with targeted deletion of FGFR1 expression in the metanephric mesenchyme. Basal serum phosphorus levels and renal cortical brush-border membrane (BBM) NaPi-2a and NaPi-2c expression were comparable between FGFR1(-/-), FGFR3(-/-), and FGFR4(-/-) mice and their wild-type counterparts. Administration of FGF23 to FGFR3(-/-) mice induced hypophosphatemia in these mice (8.0 +/- 0.4 vs. 5.4 +/- 0.3 mg/dl; p < or = 0.001) and a decrease in renal BBM NaPi-2a and NaPi-2c protein expression. Similarly, in FGFR4(-/-) mice, administration of FGF23 caused a small but significant decrease in serum phosphorus levels (8.7 +/- 0.3 vs. 7.6 +/- 0.4 mg/dl; p < or = 0.001) and in renal BBM NaPi-2a and NaPi-2c protein abundance. In contrast, injection of FGF23 into FGFR1(-/-) mice had no effects on serum phosphorus levels (5.6 +/- 0.3 vs. 5.2 +/- 0.5 mg/dl) or BBM NaPi-2a and NaPi-2c expression. These data show that FGFR1 is the predominant receptor for the hypophosphatemic action of FGF23 in vivo, with FGFR4 likely playing a minor role.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Hipofosfatemia/sangue , Túbulos Renais Proximais/metabolismo , Fósforo/sangue , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIc/metabolismo , Animais , Calcitriol/sangue , Regulação para Baixo , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/administração & dosagem , Humanos , Injeções Intraperitoneais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microvilosidades/metabolismo , Hormônio Paratireóideo/sangue , RNA Mensageiro/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/deficiência , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/metabolismo , Proteínas Recombinantes/metabolismo
12.
Am J Physiol Cell Physiol ; 297(3): C516-25, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19553564

RESUMO

The main nonhormonal mechanism for controlling inorganic phosphate (P(i)) homeostasis is renal adaptation of the proximal tubular P(i) transport rate to changes in dietary phosphate content. Opossum kidney (OK) cell line is an in vitro renal model that maintains the ability of renal adaptation to the extracellular P(i) concentration. We have studied how two competitive inhibitors of P(i) transport, arsenate [As(V)] and phosphonoformate (PFA), affect adaptation to low and high P(i) concentrations. OK cells show very high affinity for As(V) (inhibitory constant, K(i) 0.12 mM) when compared with the rat kidney. As(V) very efficiently reversed the adaptation of OK cells to low P(i) (0.1 mM), whereas PFA induced adaptation similar to 0.1 mM P(i). Adaptation with 2 mM P(i) or As(V) was characterized by decreases in the maximal velocity (V(max)) of P(i) transport and an abundance of the NaPi-IIa P(i) transporter in the plasma membrane, shown by the protein biotinylation. Conversely, PFA and 0.1 mM P(i) increased the V(max) and transporter abundance. Changes in the V(max) were limited to a 50% variation, which was not paralleled by changes in the concentration of P(i) or of the inhibitor. OK cells are very sensitive to As(V), but the effects are reversible and noncytotoxic. These effects can be interpreted as As(V) being transported into the cell, thereby mimicking a high P(i) concentration. PFA blocks the uptake of P(i) but is not transported, and it therefore simulates a low P(i) concentration inside the cell. To conclude, a mathematical definition of the adaptation process is reported, thereby explaining the limited changes in P(i) transport V(max).


Assuntos
Arseniatos/farmacologia , Foscarnet/farmacologia , Rim/citologia , Rim/efeitos dos fármacos , Fósforo/metabolismo , Animais , Linhagem Celular , Relação Dose-Resposta a Droga , Oócitos , Gambás , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/metabolismo , Xenopus
13.
J Comp Physiol B ; 178(5): 585-96, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18210130

RESUMO

Regulatory processes in phosphorus (P) homeostasis in small ruminants are quite different compared to monogastric animals. Adaptive responses of modulating hormones [parathyroid hormone (PTH) and calcitriol] to feeding variable amounts of P are lacking. Therefore, the aim of this study was to examine the influence of high dietary P intake (control diet: 4 g kg(-1) dry matter; high-P diet: 8 g kg(-1) dry matter) on the expression levels of PTH receptor (PTHR), vitamin D receptor (VDR) and Na+-dependent Pi transporters (NaPi II) in kidney and jejunum of goats starting rumination. After 3 months of feeding, plasma phosphate (Pi) and PTH concentrations were increased in the high-P diet group, whereas calcium and calcitriol were not changed. The intestinal Na+-dependent Pi transport capacity was not influenced by a high-P diet and the expression of jejunal VDR, PTHR and NaPi IIb was not modified. Interestingly, renal Na+-dependent Pi transport capacity was significantly reduced and concomitantly the expression of PTHR and NaPi IIa was decreased. In conclusion, the adaptive response of renal Pi reabsorption in goats, which were in transition from non-ruminant to ruminant stage was comparable to that of monogastric animals. In contrast, the modulation of the intestinal Pi absorption was like in adult ruminants.


Assuntos
Cabras/fisiologia , Homeostase/fisiologia , Hormônios/fisiologia , Fósforo/metabolismo , Fosfatase Alcalina/metabolismo , Animais , Peso Corporal , Calcitriol/sangue , Cálcio/sangue , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Sistema Digestório/efeitos dos fármacos , Sistema Digestório/crescimento & desenvolvimento , Sistema Digestório/metabolismo , Feminino , Expressão Gênica/efeitos dos fármacos , Cabras/crescimento & desenvolvimento , Cabras/metabolismo , Hormônios/sangue , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Jejuno/efeitos dos fármacos , Jejuno/metabolismo , Córtex Renal/efeitos dos fármacos , Córtex Renal/metabolismo , Masculino , Hormônio Paratireóideo/sangue , Fósforo/sangue , Fósforo na Dieta/administração & dosagem , Fósforo na Dieta/metabolismo , Fósforo na Dieta/farmacologia , Receptor Tipo 1 de Hormônio Paratireóideo/genética , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo , Receptores de Calcitriol/genética , Receptores de Calcitriol/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 , ATPase Trocadora de Sódio-Potássio/metabolismo
14.
Kidney Int ; 73(4): 456-64, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18046316

RESUMO

Despite similar molecular structures, the growth-related sodium/phosphate cotransporter NaPiIIc is regulated differently than the main NaPiIIa phosphate transporter. Using two-hybrid systems and immunoprecipitation, we identified several proteins that interact with NaPiIIc that might account for this differential regulation. NaPiIIc interacted with the PDZ domain-containing sodium-hydrogen exchange-regulating factor (NHERF) 1 and NHERF3 through novel binding motifs in its C terminus. NaPiIIc from brush-border membranes coprecipitated with both NHERF1 and NHERF3, with more NHERF3 co-precipitated in rats fed a low-phosphorus diet. NaPiIIc colocalizes with both NHERF1 and NHERF3 in brush-border membranes of rats fed either a low- or high-phosphorus diet. When mouse NaPiIIc was transfected into opossum kidney cells, it was localized mainly in apical microvilli and the trans-Golgi. Both confocal and total internal reflection microscopy show that NaPiIIc colocalizes with NHERF1 and NHERF3 in the apical microvilli, and this was not altered by truncation of the last three amino acids of NaPiIIc. Interactions of NaPiIIc with NHERF1 and NHERF3 were modulated by the membrane-associated 17 kDa protein (MAP17) similarly to NaPiIIa, but only the MAP17-NaPiIIc-NHERF3 complexes were internalized to the trans-Golgi. Our study shows that NaPiIIc interacts with a limited number of PDZ domain proteins, and the mechanisms and consequences of such interactions differ from those of NaPiIIa.


Assuntos
Córtex Renal/metabolismo , Domínios PDZ , Fosfoproteínas/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIc/metabolismo , Animais , Dieta , Biblioteca Gênica , Imunoprecipitação , Córtex Renal/química , Proteínas de Membrana/metabolismo , Camundongos , Fosfoproteínas/análise , Fosfoproteínas/genética , Fósforo/administração & dosagem , Mapeamento de Interação de Proteínas , Ratos , Trocadores de Sódio-Hidrogênio/análise , Trocadores de Sódio-Hidrogênio/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIc/análise , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIc/genética , Transfecção , Técnicas do Sistema de Duplo-Híbrido
15.
Cell Physiol Biochem ; 20(1-4): 99-108, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17595520

RESUMO

In mineralising tissues such as growth plate cartilage extracellular organelles derived from the chondrocyte membrane are present. These matrix vesicles (MV) possess membrane transporters that accumulate Ca(2+) and inorganic phosphate (P(i)), and initiate the formation of hydroxyapatite crystals. MV are also present in articular cartilage, and hydroxyapatite crystals are believed to promote cartilage degradation in osteoarthritic joints. In the present study, P(i) transport pathways in isolated bovine articular chondrocytes have been characterised. P(i) uptake was temperature-sensitive and could be resolved into Na(+)-dependent and Na(+)-independent components. The Na(+)-dependent component saturated at high concentrations of extracellular P(i), with a K(m) for P(i) of 0.17 mM. In solutions lacking Na(+), uptake did not fully saturate, implying that under these conditions carrier-mediated uptake is supplemented by a diffusive pathway. Both Na(+)-dependent and Na(+)-independent components were sensitive to the P(i) transport inhibitors phosphonoacetate and arsenate, although a fraction of Na(+)-independent P(i) uptake was resistant to these anions. Total P(i) uptake was optimal at pH 7.4, and reduced as pH was made more acidic or more alkaline, an effect that represented reduced Na(+)-dependent influx. RT-PCR analysis confirmed that two members of the NaPi III family, Pit-1 and Pit-2, are expressed, but that NaPi II transporters are not.


Assuntos
Condrócitos/metabolismo , Fosfatos/metabolismo , Animais , Arseniatos/farmacologia , Sequência de Bases , Transporte Biológico Ativo/efeitos dos fármacos , Cartilagem Articular/citologia , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/metabolismo , Bovinos , Condrócitos/efeitos dos fármacos , Primers do DNA/genética , Concentração de Íons de Hidrogênio , Técnicas In Vitro , Cinética , Masculino , Proteínas de Transporte de Fosfato/metabolismo , Ácido Fosfonoacéticos/farmacologia , RNA/genética , RNA/metabolismo , Sódio/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/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
16.
Cell Physiol Biochem ; 19(1-4): 43-56, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17310099

RESUMO

During calcification of bone, large amounts of phosphate (P(i)) must be transported from the circulation to the osteoid. Likely candidates for osteoblast P(i) transport are the type II sodium-phosphate cotransporters NaPi-IIa and NaPi-IIb that facilitate transcellular P(i) flux in kidney and intestine, respectively. We have therefore determined the 'cotransporters' expression in osteoblast-like cells. We have also studied the 'cotransporters' regulation by P(i) and during mineralization in vitro. Phosphate uptake and cotransporter protein expression was investigated at early, late and mineralizing culture stages of mouse (MC3T3-E1) and rat (UMR-106) osteoblast-like cells. Both NaPi-IIa and NaPi-IIb were expressed by both osteoblast-like cell lines. NaPi-IIa was upregulated in both cell lines one week after confluency. After 7 days in 3mM P(i) NaPi-IIa was strongly upregulated in both cell lines. NaPi-IIb expression was unaffected by both culture stage and P(i) supplementation. The expression of both cotransporters was unaffected by P(i) deprivation. In vitro mineralization at 1.5mM P(i) was preceded by a three-fold increase in osteoblast sodium-dependent P(i) uptake and a corresponding upregulation of both NaPi-IIa and NaPi-IIb. Their expression thus seem regulated by phosphate in a manner consistent with their playing a role in transcellular P(i) flux during mineralization.


Assuntos
Calcificação Fisiológica/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Fosfatos/farmacologia , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/metabolismo , Animais , Linhagem Celular , Regulação da Expressão Gênica , Camundongos , Osteoblastos/metabolismo , Fosfatos/farmacocinética , Fósforo na Dieta/farmacologia , Ratos , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III
17.
Toxicol Appl Pharmacol ; 214(2): 166-77, 2006 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16476458

RESUMO

Although uranium is a well-characterized nephrotoxic agent, very little is known at the cellular and molecular level about the mechanisms underlying the uptake and toxicity of this element in proximal tubule cells. The aim of this study was thus to characterize the species of uranium that are responsible for its cytotoxicity and define the mechanism which is involved in the uptake of the cytotoxic fraction of uranium using two cell lines derived from kidney proximal (LLC-PK(1)) and distal (MDCK) tubule as in vitro models. Treatment of LLC-PK(1) cells with colchicine, cytochalasin D, concanavalin A and PMA increased the sodium-dependent phosphate co-transport and the cytotoxicity of uranium. On the contrary, replacement of the extra-cellular sodium with N-methyl-D-glucamine highly reduced the transport of phosphate and the cytotoxic effect of uranium. Uranium cytotoxicity was also dependent upon the extra-cellular concentration of phosphate and decreased in a concentration-dependent manner by 0.1-10 mM phosphonoformic acid, a competitive inhibitor of phosphate uptake. Consistent with these observations, over-expression of the rat proximal tubule sodium-dependent phosphate co-transporter NaPi-IIa in stably transfected MDCK cells significantly increased the cytotoxicity of uranium, and computer modeling of uranium speciation showed that uranium cytotoxicity was directly dependent on the presence of the phosphate complexes of uranyl UO(2)(PO(4))(-) and UO(2)(HPO(4))(aq). Taken together, these data suggest that the cytotoxic fraction of uranium is a phosphate complex of uranyl whose uptake is mediated by a sodium-dependent phosphate co-transporter system.


Assuntos
Fosfatos/fisiologia , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/fisiologia , Urânio/toxicidade , Animais , Cádmio/toxicidade , Cloreto de Cálcio/farmacologia , Carbonatos/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Colchicina/farmacologia , Simulação por Computador , Concanavalina A/farmacologia , Citocalasina D/farmacologia , Dexametasona/farmacologia , Relação Dose-Resposta a Droga , Foscarnet/farmacologia , Indóis/farmacologia , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/metabolismo , Células LLC-PK1 , Maleimidas/farmacologia , Meglumina/análogos & derivados , Meglumina/farmacologia , Fosfatos/antagonistas & inibidores , Fosfatos/farmacologia , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/metabolismo , Suínos , Acetato de Tetradecanoilforbol/antagonistas & inibidores , Acetato de Tetradecanoilforbol/farmacologia , Compostos de Urânio
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA