Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Nutr ; 150(8): 2070-2076, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32470983

RESUMO

BACKGROUND: Dietary supplemental nicotinamide is used to treat hyperphosphatemia in humans. However, the mechanisms of its impact on body phosphorus homeostasis remain unclear. OBJECTIVE: This study was to determine effects and molecular mechanisms of 3 dietary nicotinamide concentrations on body phosphorus homeostasis in laying hens. METHODS: Hy-Line Brown layers (total = 21; 40 wk old; body weight: 1,876 ± 24 g) were individually housed (n = 7) and fed a corn-soybean meal-based diet supplemented with nicotinamide at 20 (N20), 140 (N140), and 1000 (N1000) mg/kg for 21 d. Serum phosphorus and fibroblast growth factor 23 (FGF23) concentrations, phosphorus and calcium excretion, and mRNA and/or protein of type II sodium-phosphate co-transporters (NPt2a, NPt2ab) and FGF23 and FGF23 receptors were measured in the intestines, calvaria, kidney, and liver. RESULTS: Hens in the N1000 group had a 16% lower serum phosphorus concentration and 22% greater phosphorus excretion than those in the N20 or N140 group (P ≤ 0.05). Compared with hens in the N20 group, hens in the N140 and N1000 groups, which did not differ, had 15-21% lower serum FGF23 concentrations, 19-22% greater calcium excretion, 43-56% lower ileum NPT2b protein production, and 1.5- to 1.6-fold greater kidney NPT2a protein production, respectively (all differences at P ≤ 0.05). CONCLUSIONS: Supplementing high concentrations of nicotinamide in diets for laying hens led to accelerated phosphorus and calcium excretions and decreased serum phosphorus and FGF23 concentrations, which were associated with downregulated intestinal NPt2b protein production. Our findings exclude kidney NPt2a protein production as a primary mechanism for the nicotinamide-induced body phosphorus loss.


Assuntos
Galinhas , Regulação da Expressão Gênica/efeitos dos fármacos , Niacinamida/farmacologia , Fósforo/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/metabolismo , Ração Animal/análise , Fenômenos Fisiológicos da Nutrição Animal , Animais , Dieta/veterinária , Suplementos Nutricionais , Relação Dose-Resposta a Droga , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Glucuronidase/genética , Glucuronidase/metabolismo , Intestinos/efeitos dos fármacos , Intestinos/fisiologia , Rim/efeitos dos fármacos , Rim/metabolismo , Proteínas Klotho , Niacinamida/administração & dosagem , Oviposição , Receptores de Fatores de Crescimento de Fibroblastos/genética , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/genética
2.
Pflugers Arch ; 471(1): 175-184, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30511265

RESUMO

Under normal physiological condition, the biomineralization process is limited to skeletal tissues and teeth and occurs throughout the individual's life. Biomineralization is an actively regulated process involving the progressive mineralization of the extracellular matrix secreted by osteoblasts in bone or odontoblasts and ameloblasts in tooth. Although the detailed molecular mechanisms underlying the formation of calcium-phosphate apatite crystals are still debated, it is suggested that calcium and phosphate may need to be transported across the membrane of the mineralizing cell, suggesting a pivotal role of phosphate transporters in bone and tooth mineralization. In this context, this short review describes the current knowledge on the role of Slc34 Na+-phosphate transporters in skeletal and tooth mineralization.


Assuntos
Osso e Ossos/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/genética , Dente/metabolismo , Animais , Biomineralização , Humanos , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/metabolismo
3.
Pflugers Arch ; 471(1): 15-42, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30244375

RESUMO

The expression cloning some 25 years ago of the first member of SLC34 solute carrier family, the renal sodium-coupled inorganic phosphate cotransporter (NaPi-IIa) from rat and human tissue, heralded a new era of research into renal phosphate handling by focussing on the carrier proteins that mediate phosphate transport. The cloning of NaPi-IIa was followed by that of the intestinal NaPi-IIb and renal NaPi-IIc isoforms. These three proteins constitute the main secondary-active Na+-driven pathways for apical entry of inorganic phosphate (Pi) across renal and intestinal epithelial, as well as other epithelial-like organs. The key role these proteins play in mammalian Pi homeostasis was revealed in the intervening decades by numerous in vitro and animal studies, including the development of knockout animals for each gene and the detection of naturally occurring mutations that can lead to Pi-handling dysfunction in humans. In addition to characterising their physiological regulation, research has also focused on understanding the underlying transport mechanism and identifying structure-function relationships. Over the past two decades, this research effort has used real-time electrophysiological and fluorometric assays together with novel computational biology strategies to develop a detailed, but still incomplete, understanding of the transport mechanism of SLC34 proteins at the molecular level. This review will focus on how our present understanding of their molecular mechanism has evolved in this period by highlighting the key experimental findings.


Assuntos
Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/metabolismo , Animais , Humanos , Transporte de Íons , Potenciais da Membrana , Domínios Proteicos , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/química , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/genética
4.
Pflugers Arch ; 471(1): 193-212, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30542786

RESUMO

Teleost fish are excellent models to study the phylogeny of the slc34 gene family, Slc34-mediated phosphate (Pi) transport and how Slc34 transporters contribute Pi homeostasis. Fish need to accumulate Pi from the diet to sustain growth. Much alike in mammals, intestinal uptake in fish is partly a paracellular and partly a Slc34-mediated transcellular process. Acute regulation of Pi balance is achieved in the kidney via a combination of Slc34-mediated secretion and/or reabsorption. A great plasticity is observed in how various species perform and combine the different processes of secretion and reabsorption. A reason for this diversity is found in one or two whole genome duplication events followed by potential gene loss; consequently, teleosts exhibit distinctly different repertoires of Slc34 transporters. Moreover, due to habitats with vastly different salinity, teleosts face the challenge of either preserving water in a hyperosmotic environment (seawater) or excreting water in hypoosmotic freshwater. An additional challenge in understanding teleost Pi homeostasis are the genome duplication and retention events that diversified peptide hormones such as parathyroid hormone and stanniocalcin. Dietary Pi and non-coding RNAs also regulate the expression of piscine Slc34 transporters. The adaptive responses of teleost Slc34 transporters to e.g. Pi diets and vitamin D are informative in the context of comparative physiology, but also relevant in applied physiology and aquaculture. In fact, Pi is essential for teleost fish growth but it also exerts significant adverse consequences if over-supplied. Thus, investigating Slc34 transporters helps tuning the physiology of commercially valuable teleost fish in a confined environment.


Assuntos
Proteínas de Peixes/metabolismo , Peixes/metabolismo , Fosfatos/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/metabolismo , Animais , Proteínas de Peixes/genética , Peixes/classificação , Peixes/genética , Filogenia , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/genética
5.
Pflugers Arch ; 471(1): 137-148, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30542787

RESUMO

The Na+-dependent phosphate transporter NaPi-IIa (SLC34A1) is mostly expressed in kidney, whereas NaPi-IIb (SLC34A2) has a wider tissue distribution with prominent expression in the lung and small intestine. NaPi-IIa is involved in renal reabsorption of inorganic phosphate (Pi) from urine, and patients with biallelic inactivating mutations in SLC34A1 develop hypophosphatemia, hypercalcemia, hypercalciuria and nephrocalcinosis, and nephrolithiasis in early childhood. Monoallelic mutations are frequent in the general population and may impact on the risk to develop kidney stones in adulthood. SNPs in close vicinity to the SLC34A1 locus associate with the risk to develop CKD. NaPi-IIb mediates high-affinity transport of Pi from the diet and appears to be mostly important during low Pi availability. Biallelic inactivating SLC34A2 mutations are found in patients with pulmonary alveolar microlithiasis, a lung disease characterized by the deposition of microcrystals. In contrast, no evidence for disturbed systemic Pi homeostasis has been reported in these patients to date. Nevertheless, NaPi-IIb-mediated intestinal Pi absorption may be a target for pharmaceutical interventions in patients with chronic kidney disease and Pi overload.


Assuntos
Doenças Inflamatórias Intestinais/etiologia , Nefropatias/etiologia , Mutação , Neoplasias/etiologia , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/genética , Animais , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Humanos , Hormônio Paratireóideo/genética , Hormônio Paratireóideo/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/metabolismo
6.
Pflugers Arch ; 471(1): 43-52, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30175376

RESUMO

Progress towards understanding the molecular mechanisms of phosphate homeostasis through sodium-dependent transmembrane uptake has long been stymied by the absence of structural information about the NaPi-II sodium-phosphate transporters. For many other coupled transporters, even those unrelated to NaPi-II, internal repeated elements have been revealed as a key feature that is inherent to their function. Here, we review recent structure prediction studies for NaPi-II transporters. Attempts to identify structural templates for NaPi-II transporters have leveraged the structural repeat perspective to uncover an otherwise obscured relationship with the dicarboxylate-sodium symporters (DASS). This revelation allowed the prediction of three-dimensional structural models of human NaPi-IIa and flounder NaPi-IIb, whose folds were evaluated by comparison with available biochemical data outlining the transmembrane topology and solvent accessibility of various regions of the protein. Using these structural models, binding sites for sodium and phosphate were proposed. The predicted sites were tested and refined based on detailed electrophysiological and biochemical studies and were validated by comparison with subsequently reported structures of transporters belonging to the AbgT family. Comparison with the DASS transporter VcINDY suggested a conformational mechanism involving a large, two-domain structural change, known as an elevator-like mechanism. These structural models provide a foundation for further studies into substrate binding, conformational change, kinetics, and energetics of sodium-phosphate transport. We discuss future opportunities, as well as the challenges that remain.


Assuntos
Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/química , Substituição de Aminoácidos , Animais , Humanos , Simulação de Dinâmica Molecular , Fosfatos/metabolismo , Sódio/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/metabolismo
7.
Pflugers Arch ; 471(1): 165-173, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30343332

RESUMO

There has recently been significant interest in the concept of directly targeting intestinal phosphate transport to control hyperphosphatemia in patients with chronic kidney disease. However, we do not have a complete understanding of the cellular mechanisms that govern dietary phosphate absorption. Studies in the 1970s documented both active and passive pathways for intestinal phosphate absorption. However, following the cloning of the intestinal SLC34 cotransporter, NaPi-IIb, much of the research focused on the role of this protein in active transcellular phosphate absorption and the factors involved in its regulation. Generation of a conditional NaPi-IIb knockout mouse has demonstrated that this protein is critical for the maintenance of skeletal integrity during periods of phosphate restriction and that under normal physiological conditions, the passive sodium-independent pathway is likely be the more dominant pathway for intestinal phosphate absorption. The review aims to summarise the most recent developments in our understanding of the role of the intestine in phosphate homeostasis, including the acute and chronic renal adaptations that occur in response to dietary phosphate intake. Evidence regarding the overall contribution of the transcellular and paracellular pathways for phosphate absorption will be discussed, together with the clinical benefit of inhibiting these pathways for the treatment of hyperphosphatemia in chronic kidney disease.


Assuntos
Absorção Intestinal , Fosfatos/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/metabolismo , Animais , Homeostase , Humanos , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/genética
8.
Pflugers Arch ; 471(1): 67-82, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30022249

RESUMO

The sorting and stabilization of proteins at specific subcellular domains depend upon the formation of networks build up by specific protein-protein interactions. In addition, protein networks also ensure the specificity of many regulatory processes by bringing together regulatory molecules with their targets. Whereas the success on the identification of protein-protein interactions is (up to a point) technology-driven, the assignment of functional roles to specific partners remains a major challenge. This review summarizes the work that led to the identification of partners of the Na+/phosphate cotransporter NaPi-IIa as well as the effects of the interactions in the expression and/or regulation of the cotransporter.


Assuntos
Mapas de Interação de Proteínas , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/metabolismo , Animais , Sítios de Ligação , Humanos , Túbulos Renais/metabolismo , Ligação Proteica , Reabsorção Renal , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/química , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/genética
9.
Tsitol Genet ; 49(4): 11-6, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26419064

RESUMO

Cichorium intybus L. is an important vegetable crop used as salad (leaf form) and for the production of coffee substitutes (root form). At the same time these plants can also be used in biotechnologies for synthesis of pharmaceutical proteins. Here we report the possibility of high frequency Agrobacterium rhizogenes- or A. tumefaciens-mediated transformation of C. intybus L. for construction of transgenic "hairy" roots and plants. The used plasmids contained target human interferonifn-α2b gene, Mycobacterium tuberculosis ESAT6:Ag85B antigene esxA::fbpB(ΔTMD) fused gene and human telomerase reverse transcriptase h Tert gene. Using of nptII gene as a selective one was preferable to the bar gene for chicory. In this case the frequency of transgenic plants or "hairy" roots formation was significantly higher. Cultivation of explants on the medium with Basta in concentration 1-2 mg/l have led to plants death or to significant reduction of number of shoots formed. Frequency of "hairy" roots formation varied from 5.9 to 42.3% after A. rhizogenes-mediated transformation. Frequency of regeneration of transgenic plants varied from 10 to 86% after A. tumefaciens-mediated transformation. Both A. rhizogenes- and A. tumefaciens-mediated transformation frequency depended on the type of explants, roots or cotyledons, and vector used. Usage of A. tumefaciens carrying pCB064 plasmid (target esxA:fbpB(ΔTMD) fused gene and nptII selective gene) resulted in the most effective regeneration of transgenic plants with regeneration frequency up to 86%. In the case of chicory A. rhizogenes-mediated transformation the highest regeneration frequency up to 42.3% was demonstrated using p CB161 vector with ifn-α2b target gene and nptII selective gene.


Assuntos
Agrobacterium/genética , Cichorium intybus/genética , Cotilédone/genética , Raízes de Plantas/genética , Plasmídeos/metabolismo , Transformação Genética , Aciltransferases/genética , Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Cichorium intybus/anatomia & histologia , Cotilédone/anatomia & histologia , Marcadores Genéticos , Vetores Genéticos , Interferon-alfa/genética , Mycobacterium tuberculosis/química , Raízes de Plantas/anatomia & histologia , Plantas Geneticamente Modificadas , Plasmídeos/química , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/genética , Telomerase/genética
10.
Urolithiasis ; 43 Suppl 1: 65-76, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25096800

RESUMO

Genetically engineered mouse models (GEMMs) have been highly instrumental in elucidating gene functions and molecular pathogenesis of human diseases, although their use in studying kidney stone formation or nephrolithiasis remains relatively limited. This review intends to provide an overview of several knockout mouse models that develop interstitial calcinosis in the renal papillae. Included herein are mice deficient for Tamm-Horsfall protein (THP; also named uromodulin), osteopontin (OPN), both THP and OPN, Na(+)-phosphate cotransporter Type II (Npt2a) and Na(+)/H(+) exchanger regulatory factor (NHERF-1). The baseline information of each protein is summarized, along with key morphological features of the interstitial calcium deposits in mice lacking these proteins. Attempts are made to correlate the papillary interstitial deposits found in GEMMs with Randall's plaques, the latter considered precursors of idiopathic calcium stones in patients. The pathophysiology that underlies the renal calcinosis in the knockout mice is also discussed wherever information is available. Not all the knockout models are allocated equal space because some are more extensively characterized than others. Despite the inroads already made, the exact physiological underpinning, origin, evolution and fate of the papillary interstitial calcinosis in the GEMMs remain incompletely defined. Greater investigative efforts are warranted to pin down the precise role of the papillary interstitial calcinosis in nephrolithiasis using the existing models. Additionally, more sophisticated, second-generation GEMMs that allow gene inactivation in a time-controlled manner and "compound mice" that bear several genetic alterations are urgently needed, in light of mounting evidence that nephrolithiasis is a multifactorial, multi-stage and polygenic disease.


Assuntos
Calcinose/genética , Nefropatias/genética , Medula Renal , Animais , Modelos Animais de Doenças , Camundongos/genética , Camundongos Knockout , Osteopontina/genética , Fosfoproteínas/genética , Trocadores de Sódio-Hidrogênio/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/genética , Uromodulina/genética
11.
Plant Cell Rep ; 34(3): 469-81, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25504050

RESUMO

KEY MESSAGE: Marker-free transgenic plants can be generated with high efficiency by using the Cre/ lox P self-excision system controlled by the pollen- and embryo-specific Arabidopsis DLL promoter. In this work, we aimed to study the feasibility of using the pollen- and embryo-specific DLL promoter of the At4g16160 gene from Arabidopsis thaliana in a Cre/loxP self-excision strategy. A Cre/loxP self-excision cassette controlled by the DLL promoter was introduced into the tobacco genome via Agrobacterium-mediated transformation. No evidence for premature activation of the Cre/loxP system was observed in primary transformants. The efficiency of nptII removal during pollen and embryo development was investigated in transgenic T1 progenies derived from eight self- and four cross-pollinated T0 lines, respectively. Segregation and rooting assays were performed to select recombined T1 plants. Molecular analyses of these plants confirmed the excision event in all analysed T0 lines and marker-free transgenic T1 plants were obtained with efficiency of up to 96.2%. The Arabidopsis DLL promoter appears to be a strong candidate to drive Cre-mediated recombination not only in tobacco as a model plant, but also in other plant species.


Assuntos
Proteínas de Arabidopsis/genética , Canais Iônicos/genética , Nicotiana/genética , Plantas Geneticamente Modificadas , Pólen/genética , Regiões Promotoras Genéticas/genética , Agrobacterium/genética , Sequência de Bases , Regulação da Expressão Gênica de Plantas , Marcadores Genéticos , Vetores Genéticos , Integrases/genética , Dados de Sequência Molecular , Sementes/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/genética , Transformação Bacteriana
12.
Appl Microbiol Biotechnol ; 99(7): 3169-77, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25515813

RESUMO

Fibroblast growth factor 23 (FGF23) is a bone-derived endocrine regulator of phosphate homeostasis and has been considered as a potential therapeutic target for hypophosphatemic disorders. Herein, we isolated a novel FGF23-binding peptide by screening a phage display library with FGF23180-205, the minimal epitope of FGF23 binding to the binary fibroblast growth factor receptor (FGFR)-Klotho complex. The corresponding peptide (referred to as 23-b6) showed high homology to the immunoglobulin-like (Ig-like) domain III (D3) of FGFR1c, the predominant receptor mediating the phosphaturic activity of FGF23. The 23-b6 peptide and panning target FGF23180-205 carried opposite charges and shared similar hydrophilic profiles. Functional analysis indicated that synthetic 23-b6 peptide exhibited antagonistic effect on the inhibition of phosphate uptake by FGF23 in opossum kidney cells (OK cells). The mechanisms of 23-b6 peptide impairing the bioactivity of FGF23 involved blockade of the activation of Erk cascade and up-regulation of NaPi-2a and NaPi-2c expression in OK cells. Our results demonstrate that the 23-b6 peptide is a potent FGF23 antagonist with increased effect on phosphate uptake in kidney cells and might have therapeutic potentials in hypophosphatemic disorders characterized by an abnormally high level of FGF23.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Biblioteca de Peptídeos , Peptídeos/farmacologia , Fosfatos/farmacocinética , Animais , Células Cultivadas/efeitos dos fármacos , Epitopos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Fatores de Crescimento de Fibroblastos/farmacologia , Glucuronidase/metabolismo , Rim/citologia , Proteínas Klotho , Gambás , Peptídeos/metabolismo , Fosfatos/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/metabolismo
13.
Biophys J ; 106(6): 1268-79, 2014 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-24655502

RESUMO

Phosphate plays essential biological roles and its plasma level in humans requires tight control to avoid bone loss (insufficiency) or vascular calcification (excess). Intestinal absorption and renal reabsorption of phosphate are mediated by members of the SLC34 family of sodium-coupled transporters (NaPi-IIa,b,c) whose membrane expression is regulated by various hormones, circulating proteins, and phosphate itself. Consequently, NaPi-II proteins are also potentially important pharmaceutical targets for controlling phosphate levels. Their crucial role in Pi homeostasis is underscored by pathologies resulting from naturally occurring SLC34 mutations and SLC34 knockout animals. SLC34 isoforms have been extensively studied with respect to transport mechanism and structure-function relationships; however, the three-dimensional structure is unknown. All SLC34 transporters share a duplicated motif comprising a glutamine followed by a stretch of threonine or serine residues, suggesting the presence of structural repeats as found in other transporter families. Nevertheless, standard bioinformatic approaches fail to clearly identify a suitable template for molecular modeling. Here, we used hydrophobicity profiles and hidden Markov models to define a structural repeat common to all SLC34 isoforms. Similar approaches identify a relationship with the core regions in a crystal structure of Vibrio cholerae Na(+)-dicarboxylate transporter VcINDY, from which we generated a homology model of human NaPi-IIa. The aforementioned SLC34 motifs in each repeat localize to the center of the model, and were predicted to form Na(+) and Pi coordination sites. Functional relevance of key amino acids was confirmed by biochemical and electrophysiological analysis of expressed, mutated transporters. Moreover, the validity of the predicted architecture is corroborated by extensive published structure-function studies. The model provides key information for elucidating the transport mechanism and predicts candidate substrate binding sites.


Assuntos
Simulação de Dinâmica Molecular , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/química , Motivos de Aminoácidos , Sequência de Aminoácidos , Sítios de Ligação , Humanos , Dados de Sequência Molecular , Mutação , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/metabolismo
14.
Contrib Nephrol ; 180: 124-37, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23652555

RESUMO

Deregulated phosphate homeostasis can lead to a wide range of disorders, including myopathy, cardiac dysfunction, and skeletal abnormalities. Therefore, characterization of the molecular regulation of phosphate metabolism is of pathophysiological and clinical significance. Hyp mouse is the model for human X-linked hypophosphatemia which is due to mutations that inactivate the endopeptidases of the X chromosome (PHEX). PHEX inactivation leads to increased serum levels of fibroblast growth factor 23 (FGF23), a phosphaturic hormone that induces excessive renal phosphate excretion and severe hypophosphatemia. The expression of WNT signaling components is increased in Hyp mice. To determine the potential role of WNT signaling in FGF23-mediated hypophosphatemia, we cross-bred Hyp mice with mice deficient in the WNT coreceptor low-density lipoprotein receptor-related protein 6 (Lrp6) to generate Hyp and Lrp6 double mutant mice (Hyp/Lrp6). Like Hyp mice, Hyp/Lrp6 double mutants maintained high serum levels of FGF23, and accordingly exhibited hypophosphatemia to the same degree as the Hyp mice did, indicating that genetically reducing WNT signaling does not impact FGF23-induced phosphaturia. Moreover, similar to Hyp mice, the Hyp/Lrp6 double mutants also exhibited reduced mineralization of the bone, further supporting that reduced WNT signaling does not affect the chronic phosphate wasting caused by excess FGF23 in these mice. In further support of our finding, injection of bioactive FGF23 protein into Lrp6 mutant mice reduced serum phosphate levels to a similar degree as FGF23 injection into wild-type mice. Our in vivo studies provide genetic and pharmacological evidence for a WNT-independent function of FGF23 in the regulation of phosphate homeostasis.


Assuntos
Modelos Animais de Doenças , Raquitismo Hipofosfatêmico Familiar/fisiopatologia , Fatores de Crescimento de Fibroblastos/fisiologia , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/deficiência , Endopeptidase Neutra Reguladora de Fosfato PHEX/fisiologia , Via de Sinalização Wnt , Animais , Raquitismo Hipofosfatêmico Familiar/diagnóstico por imagem , Raquitismo Hipofosfatêmico Familiar/etiologia , Raquitismo Hipofosfatêmico Familiar/genética , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/sangue , Fatores de Crescimento de Fibroblastos/toxicidade , Homeostase , Hipofosfatemia Familiar/genética , Hipofosfatemia Familiar/metabolismo , Rim/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Masculino , Camundongos , Camundongos Knockout , Endopeptidase Neutra Reguladora de Fosfato PHEX/genética , Fosfatos/metabolismo , Radiografia , Proteínas Recombinantes/toxicidade , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/biossíntese , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/genética
15.
Pediatr Nephrol ; 27(9): 1477-87, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22350303

RESUMO

Regulation of phosphate homeostasis is critical for many biological processes, and both hypophosphatemia and hyperphosphatemia can have adverse clinical consequences. Only a very small percentage (1%) of total body phosphate is present in the extracellular fluid, which is measured by routine laboratory assays and does not reflect total body phosphate stores. Phosphate is absorbed from the gastrointestinal tract via the transcellular route [sodium phosphate cotransporter 2b (NaPi2b)] and across the paracellular pathway. Approximately 85% of the filtered phosphate is reabsorbed from the kidney, predominantly in the proximal tubule, by NaPi2a and NaPi2c, which are present on the brush border membrane. Renal phosphate transport is tightly regulated. Dietary phosphate intake, parathyroid hormone (PTH), 1,25 (OH)2 vitamin D3, and fibroblast growth factor 23 (FGF23) are the principal regulators of phosphate reabsorption from the kidney. Recent advances in genetic techniques and animal models have identified many genetic disorders of phosphate homeostasis. Mutations in NaPi2a and NaPi2c; and hormonal dysregulation of PTH, FGF23, and Klotho, are primarily responsible for most genetic disorders of phosphate transport. The main focus of this educational review article is to discuss the genetic and clinical features of phosphate regulation disorders and provide understanding and treatment options.


Assuntos
Hiperfosfatemia/genética , Hiperfosfatemia/metabolismo , Hipofosfatemia Familiar/genética , Hipofosfatemia Familiar/metabolismo , Fosfatos/metabolismo , Animais , Fator de Crescimento de Fibroblastos 23 , Homeostase , Humanos , Erros Inatos do Metabolismo/genética , Erros Inatos do Metabolismo/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/metabolismo
16.
Adv Chronic Kidney Dis ; 18(2): 63-76, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21406290

RESUMO

Inorganic phosphate (Pi) is essential for all living organisms. Bound to organic molecules, Pi fulfills structural, metabolic, and signaling tasks. Therefore, cell growth and maintenance depends on efficient transport of Pi across cellular membranes into the intracellular space. Uptake of Pi requires energy because the substrate is transported against its electrochemical gradient. Till recently, 2 major families of physiologically relevant Pi-specific transporters have been identified: the solute carrier families Slc34 and Slc20. Interestingly, phylogenetic links can be detected between prokaryotic and eukaryotic transporters in both families. Because less complex model organisms are often instrumental in establishing paradigms for protein function in human beings, a brief assessment of Slc34 and Slc20 phylogeny is of interest.


Assuntos
Absorção Intestinal , Rim/metabolismo , Fosfatos/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/metabolismo , Absorção , Transporte Biológico Ativo , Homeostase , Humanos , Transporte de Íons , Cinética , Filogenia , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/genética , Relação Estrutura-Atividade
17.
Bone ; 45 Suppl 1: S2-7, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19232403

RESUMO

The renal type II Na/Pi cotransporters, Na/Pi-IIa and Na/Pi-IIc, are expressed in the brush border membrane (BBM) of the renal proximal tubule cells. Because it has long been thought that Na/Pi-IIa alone can regulate the reabsorption of phosphate in the proximal renal tubules, Na/Pi-IIc has not been paid much attention by the renal research community. Recent studies, however, have identified Na/Pi-IIc mutations as the defective cause of hereditary hypophosphatemic rickets with hypercalciuria (HHRH). This finding indicates that Na/Pi-IIc has a rather important role in renal Pi reabsorption and bone mineralization, and that it may be a key determinant of plasma Pi concentrations in humans. Studies of Na/Pi-IIc mice indicate that Na/Pi-IIc is necessary for normal calcium homeostasis, but its role in the regulation of Pi metabolism and bone physiology may be different from that in HHRH patients. Of note, Na/Pi-IIc KO mice display abnormal vitamin D regulation without hypophosphatemia or hyperphosphaturia. Thus, Na/Pi-IIc may be involved in regulating renal vitamin D synthesis in the proximal tubular cells. The identification of proteins that interact with Na/Pi-IIc is an important area of future research. The physiologic roles of Na/Pi-IIa and Na/Pi-IIc require future elucidation.


Assuntos
Fosfatos/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/metabolismo , Animais , Raquitismo Hipofosfatêmico Familiar/complicações , Raquitismo Hipofosfatêmico Familiar/metabolismo , Humanos , Hipercalciúria/complicações , Hipercalciúria/metabolismo , Mutação/genética , Fosfoproteínas/genética , Trocadores de Sódio-Hidrogênio/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/química , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/genética
18.
J Membr Biol ; 215(2-3): 81-92, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17443384

RESUMO

The temperature dependence of the transport kinetics of flounder Na(+)-coupled inorganic phosphate (P(i)) cotransporters (NaPi-IIb) expressed in Xenopus oocytes was investigated using radiotracer and electrophysiological assays. (32)P(i) uptake was strongly temperature-dependent and decreased by approximately 80% at a temperature change from 25 degrees C to 5 degrees C. The corresponding activation energy (E (a)) was approximately 14 kcal mol(-1) for the cotransport mode. The temperature dependence of the cotransport and leak modes was determined from electrogenic responses to 1 mM P(i) and phosphonoformic acid (PFA), respectively, under voltage clamp. The magnitude of the P(i)- and PFA-induced changes in holding current decreased with temperature. E (a) at -100 mV for the cotransport and leak modes was approximately 16 kcal mol(-1) and approximately 11 kcal mol(-1), respectively, which suggested that the leak is mediated by a carrier, rather than a channel, mechanism. Moreover, E (a) for cotransport was voltage-independent, suggesting that a major conformational change in the transport cycle is electroneutral. To identify partial reactions that confer temperature dependence, we acquired presteady-state currents at different temperatures with 0 mM P(i) over a range of external Na(+). The relaxation time constants increased, and the peak time constant shifted toward more positive potentials with decreasing temperature. Likewise, there was a depolarizing shift of the charge distribution, whereas the total available charge and apparent valency predicted from single Boltzmann fits were temperature-independent. These effects were explained by an increased temperature sensitivity of the Na(+)-debinding rate compared with the other voltage-dependent rate constants.


Assuntos
Proteínas de Peixes/fisiologia , Linguado/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/fisiologia , Animais , Eletrofisiologia , Feminino , Proteínas de Peixes/genética , Proteínas de Peixes/metabolismo , Linguado/genética , Foscarnet/farmacologia , Cinética , Potenciais da Membrana/efeitos dos fármacos , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Oócitos/fisiologia , Técnicas de Patch-Clamp , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/metabolismo , Temperatura , Xenopus laevis
19.
Plant Cell Rep ; 26(7): 1035-44, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17333020

RESUMO

A selectable marker gene (SMG), usually conferring resistance to an antibiotic or herbicide, is generally introduced into the plant cells with the gene(s) for the trait of interest to allow only the cells that have integrated and express the foreign sequences to regenerate into a plant. The availability of several SMGs for each plant species is useful for both basic and applied research to combine several genes of interest in the same plant. A selection system based on gabaculine (3-amino-2,3-dihydrobenzoic acid) as the selective substance and the bacterial hemL gene [encoding a mutant for of the enzyme glutamate 1-semialdehyde aminotransferase (GSA-AT)] as the SMG was previously used for genetic transformation of tobacco. The hemL gene is a good candidate for a safe SMG, because GSA-AT is present in all plants and is likely involved in one metabolic step only, so that unintended effects of its overexpression in plants are not probable. In this work, we have compared this new selection system with the conventional, kanamycin-based system for alfalfa Agrobacterium-mediated transformation. The hemL and NptII genes were placed together into a T-DNA under the control of identical promoters and terminators. We show that the gabaculine-based system is more efficient than the conventional, kanamycin-based system. The inheritance of hemL was Mendelian, and no obvious phenotypic effect of its expression was observed.


Assuntos
Engenharia Genética/métodos , Medicago sativa/genética , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Seleção Genética , Ácidos Cicloexanocarboxílicos/farmacologia , Regulação da Expressão Gênica de Plantas , Marcadores Genéticos/genética , Plantas Geneticamente Modificadas , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/metabolismo , Técnicas de Cultura de Tecidos
20.
J Comp Physiol B ; 177(2): 193-203, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17053890

RESUMO

P homeostasis affected by high or low Ca and/or P supply in preruminant goats was characterized by balance studies in vivo. The main excretion pathway was the renal P(i) excretion whose extent was modulated by variations in dietary P and/or Ca supply. Faecal P excretion remained low irrespective of dietary regimen. The balance data were combined with respective in vitro data on P(i) transport properties and their adaptation in response to changes in dietary Ca and/or P intake. Therefore, P(i) transport capacities were determined by P(i) uptake into brush border membrane vesicles of jejunum and kidney. Epithelial P(i) transporters were determined semiquantitatively by northern and western blot analyses in jejunum, kidney and salivary gland. Renal P(i) transport was downregulated by doubling dietary P supply while doubling both, Ca and P as well as restrictive Ca at unchanged P led to slight, but not significant reductions in renal P(i) transport. Jejunal P(i) transport was reduced by P excess (doubling P and doubling both, Ca and P), but only NaPi IIb protein expression was significantly diminished. In conclusion, the significance of epithelial adaptation to dietary Ca and P supply for P homeostasis is discussed in preruminant goats.


Assuntos
Cálcio da Dieta/farmacologia , Cabras/metabolismo , Mucosa Intestinal/metabolismo , Fosfatos/metabolismo , Fósforo na Dieta/farmacologia , Ração Animal , Animais , Transporte Biológico/efeitos dos fármacos , Calcitriol/sangue , Cálcio/metabolismo , Regulação da Expressão Gênica , Mucosa Intestinal/citologia , Intestinos/citologia , Jejuno/metabolismo , Rim/metabolismo , Masculino , Rúmen/metabolismo , Glândulas Salivares/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo II/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...