Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 212
Filter
Add more filters










Publication year range
2.
Rev Sci Tech ; 35(2): 561-575, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27917971

ABSTRACT

Transhumant pastoralism remains a prevalent form of land use across Europe, especially in mountain areas. Besides generating food and other products, it provides a range of public goods and services that are often highly valued by broader society. But transhumance faces structural challenges associated with life in remote mountain areas, including economic pressures, lack of services, low prestige, and ageing populations. These threaten its future. The decline of transhumant systems leads to ecological, economic, and socio-cultural losses, e.g. the loss of biodiversity, of shared cultural heritage based on pastoral practices, and of common property institutions. There are a number of activities that will help to ensure the future of European transhumance, including: i) raising awareness of the public services it provides, ii) better integrating pastoral issues into national agricultural policies, and iii) increasing support for so-called high nature value (HNV) farmlands, which often feature extensive grazing.


Le pastoralisme transhumant constitue encore aujourd'hui une modalité courante d'utilisation des terres en Europe, particulièrement dans les régions montagneuses. Outre les denrées alimentaires et autres produits directs, le pastoralisme est créateur d'une panoplie de services et de biens publics auxquels la société accorde souvent une grande valeur. Mais la transhumance est également confrontée aux difficultés structurelles inhérentes à la vie dans les régions montagneuses isolées, en particulier la pression économique, la pénurie de services, le manque d'attractivité et le vieillissement de la population. L'avenir du pastoralisme s'en trouve menacé. Le déclin des systèmes de transhumance entraîne des pertes écologiques, économiques et socioculturelles, notamment en termes de biodiversité, de patrimoine culturel commun fondé sur les pratiques pastorales et d'institutions collectives. Quelques mesures pourraient contribuer à préserver l'avenir de la transhumance en Europe : i) faire prendre conscience des services que la transhumance rend à la société ; ii) intégrer davantage les problématiques du pastoralisme dans les politiques agricoles nationales ; iii) renforcer le soutien accordé aux systèmes agricoles dits à haute valeur naturelle, dont le pâturage extensif fait partie.


El pastoreo trashumante sigue siendo una forma común de utilización de la tierra en toda Europa, especialmente en las zonas montañosas. Además de generar productos alimentarios y de otra índole, la trashumancia aporta una serie de bienes y servicios públicos a los que la sociedad en general suele conceder gran valor. La trashumancia, sin embargo, afronta problemas estructurales ligados a la vida en zonas montañosas aisladas, tales como las presiones económicas, la falta de servicios, el escaso prestigio de esta actividad o el envejecimiento de las poblaciones que la ejercen. Todo ello amenaza su futuro. El declive de los sistemas trashumantes trae consigo pérdidas ecológicas, económicas y socioculturales, por ejemplo el empobrecimiento de la diversidad biológica, el de un patrimonio cultural común basado en los usos pastorales y el de las instituciones de propiedad colectiva. Entre las diversas actividades que ayudarán a asegurar un porvenir a la trashumancia europea, los autores destacan las siguientes: i) sensibilización con respecto a los servicios públicos que presta; ii) mayor integración de los asuntos relativos al pastoreo en las políticas agrícolas nacionales; y iii) mayor apoyo a las llamadas tierras agrícolas de gran valor natural (high nature value: HNV), que a menudo albergan pastizales extensivos.


Subject(s)
Animal Husbandry/methods , Animals , Europe , European Union , Humans , Policy , Seasons
3.
Compr Physiol ; 4(2): 771-92, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24715567

ABSTRACT

In the kidney, both anions, phosphate and sulfate, are almost freely filtered and afterwards reclaimed (reabsorbed) to a large extent from tubular fluid along the proximal tubules. Under normal dietary conditions, fractional excretion of these anions is approximately 10%. Reabsorption of both anions occurs along the proximal tubules by active, saturable, and regulated transepithelial processes. Most of the transporters involved in renal handling of phosphate and sulfate have been identified and their transport functions as well as their cellular localizations have been described in detail. The role of these transporters in the renal handling of phosphate and sulfate has been investigated by the use of several mice knock out models and also by analysis of several inherited human diseases. Numerous hormonal and nonhormonal factors, have been described that alter renal excretion of phosphate or sulfate by mechanisms that alter the abundance of known phosphate/sulfate transporters and consequently renal excretion. These mechanisms contribute to the homeostasis of the extracellular concentrations of phosphate and sulfate.


Subject(s)
Homeostasis/physiology , Kidney/metabolism , Phosphates/metabolism , Renal Reabsorption/physiology , Sulfates/metabolism , Animals , Humans
4.
Pflugers Arch ; 458(1): 39-52, 2009 May.
Article in English | MEDLINE | ID: mdl-18758808

ABSTRACT

Homeostasis of inorganic phosphate (P(i)) is primarily an affair of the kidneys. Reabsorption of the bulk of filtered P(i) occurs along the renal proximal tubule and is initiated by apically localized Na(+)-dependent P(i) cotransporters. Tubular P(i) reabsorption and therefore renal excretion of P(i) is controlled by a number of hormones, including phosphatonins, and metabolic factors. In most cases, regulation of P(i) reabsorption is achieved by changing the apical abundance of Na(+)/Pi cotransporters. The regulatory mechanisms involve various signaling pathways and a number of proteins that interact with Na(+)/P(i) cotransporters.


Subject(s)
Kidney Tubules, Proximal/metabolism , Phosphates/metabolism , Sodium-Phosphate Cotransporter Proteins/physiology , Acidosis/physiopathology , Animals , Cholecalciferol/physiology , Circadian Rhythm , Diet , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/physiology , Gene Expression Regulation , Glucuronidase/physiology , Gonadal Steroid Hormones/physiology , Homeostasis , Humans , Ion Transport , Klotho Proteins , Parathyroid Hormone/physiology , Phosphates/urine , Phosphoproteins/physiology , Potassium Deficiency/physiopathology , Signal Transduction , Sodium-Hydrogen Exchangers/physiology , Sodium-Phosphate Cotransporter Proteins, Type IIa/physiology , Sodium-Phosphate Cotransporter Proteins, Type IIc/physiology
5.
Kidney Int ; 72(2): 166-73, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17457376

ABSTRACT

Hyperphosphatemia is an important consequence of chronic renal failure (CRF). Lowering of the plasma phosphate concentration is believed to be critical in the management of patients with CRF, especially those on dialysis. Reports of the effect of CRF on the intestinal handling of phosphate in vitro have been conflicting; but what happens in vivo has not been studied. What effect a reduction in the dietary phosphate intake has on intestinal phosphate absorption in CRF in vivo is unclear. In this study, we have used the in situ intestine loop technique to determine intestinal phosphate absorption in the 5/6-nephrectomy rat model of CRF under conditions of normal and restricted dietary phosphate intake. In this model of renal disease, we found that there is no significant change in the phosphate absorption in either the duodenum or jejunum regardless of the dietary phosphate intake. There was also no change in the expression of the messenger RNA of the major intestinal phosphate carrier the sodium-dependent-IIb transporter. Furthermore, we found no change in the intestinal villus length or in the location of phosphate uptake along the villus. Our results indicate that in CRF, unlike the kidney, there is no reduction in phosphate transport across the small intestine. This makes intestinal phosphate absorption a potential target in the prevention and treatment of hyperphosphatemia.


Subject(s)
Hypophosphatemia/metabolism , Intestinal Absorption , Kidney Failure, Chronic/complications , Phosphates/pharmacokinetics , Animals , Disease Models, Animal , Homeostasis , Hypophosphatemia/etiology , Male , Nephrectomy , Rats , Rats, Sprague-Dawley
6.
Kidney Int ; 70(10): 1725-34, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17003823

ABSTRACT

Renal Fanconi syndrome occurs in about 1-5% of all children treated with Ifosfamide (Ifo) and impairment of renal phosphate reabsorption in about 20-30% of them. Pathophysiological mechanisms of Ifo-induced nephropathy are ill defined. The aim has been to investigate whether Ifo metabolites affect the type IIa sodium-dependent phosphate transporter (NaPi-IIa) in viable opossum kidney cells. Ifo did not influence viability of cells or NaPi-IIa-mediated transport up to 1 mM/24 h. Incubation of confluent cells with chloroacetaldehyde (CAA) and 4-hydroperoxyIfosfamide (4-OH-Ifo) led to cell death by necrosis in a concentration-dependent manner. At low concentrations (50-100 microM/24 h), cell viability was normal but apical phosphate transport, NaPi-IIa protein, and -mRNA expression were significantly reduced. Coincubation with sodium-2-mercaptoethanesulfonate (MESNA) prevented the inhibitory action of CAA but not of 4-OH-Ifo; DiMESNA had no effect. Incubation with Ifosfamide-mustard (Ifo-mustard) did alter cell viability at concentrations above 500 microM/24 h. At lower concentrations (50-100 microM/24 h), it led to significant reduction in phosphate transport, NaPi-IIa protein, and mRNA expression. MESNA did not block these effects. The effect of Ifo-mustard was due to internalization of NaPi-IIa. Cyclophosphamide-mustard (CyP-mustard) did not have any influence on cell survival up to 1000 microM, but the inhibitory effect on phosphate transport and on NaPi-IIa protein was the same as found after Ifo-mustard. In conclusion, CAA, 4-OH-Ifo, and Ifo- and CyP-mustard are able to inhibit sodium-dependent phosphate cotransport in viable opossum kidney cells. The Ifo-mustard effect took place via internalization and reduction of de novo synthesis of NaPi-IIa. Therefore, it is possible that Ifo-mustard plays an important role in pathogenesis of Ifo-induced nephropathy.


Subject(s)
Acetaldehyde/analogs & derivatives , Ifosfamide/analogs & derivatives , Ifosfamide/pharmacology , Phosphates/metabolism , Phosphoramide Mustards/pharmacology , Sodium-Phosphate Cotransporter Proteins, Type IIa/drug effects , Sodium-Phosphate Cotransporter Proteins, Type IIa/metabolism , Acetaldehyde/pharmacology , Animals , Antineoplastic Agents, Alkylating , Biological Transport/drug effects , Cell Death/drug effects , Cell Line , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Ifosfamide/metabolism , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , Mesna/pharmacology , Opossums , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIa/genetics
7.
Kidney Int ; 70(9): 1548-59, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16955105

ABSTRACT

Members of the SLC34 gene family of solute carriers encode for three Na+-dependent phosphate (P i) cotransporter proteins, two of which (NaPi-IIa/SLC34A1 and NaPi-IIc/SLC34A3) control renal reabsorption of P i in the proximal tubule of mammals, whereas NaPi-IIb/SCLC34A2 mediates P i transport in organs other than the kidney. The P i transport mechanism has been extensively studied in heterologous expression systems and structure-function studies have begun to reveal the intricacies of the transport cycle at the molecular level using techniques such as cysteine scanning mutagenesis, and voltage clamp fluorometry. Moreover, sequence differences between the three types of cotransporters have been exploited to obtain information about the molecular determinants of hormonal sensitivity and electrogenicity. Renal handling of P i is regulated by hormonal and non-hormonal factors. Changes in urinary excretion of P i are almost invariably mirrored by changes in the apical expression of NaPi-IIa and NaPi-IIc in proximal tubules. Therefore, understanding the mechanisms that control the apical expression of NaPi-IIa and NaPi-IIc as well as their functional properties is critical to understanding how an organism achieves P i homeostasis.


Subject(s)
Kidney Tubules, Proximal/metabolism , Phosphates/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIa/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIb/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIc/metabolism , Animals , Homeostasis , Humans , Mice , Parathyroid Hormone/physiology , Sodium-Phosphate Cotransporter Proteins, Type IIa/chemistry , Sodium-Phosphate Cotransporter Proteins, Type IIa/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIb/chemistry , Sodium-Phosphate Cotransporter Proteins, Type IIb/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIc/chemistry , Sodium-Phosphate Cotransporter Proteins, Type IIc/genetics , Structure-Activity Relationship
8.
Kidney Int ; 69(3): 495-503, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16514432

ABSTRACT

The major renal Na(+)/phosphate cotransporter, NaPi-IIa, is regulated by a number of factors including parathyroid hormone (PTH), dopamine, and dietary phosphate intake. PTH induces the acute internalization of NaPi-IIa from the brush border membrane (BBM) and its routing to and subsequent degradation in lysosomes. Previous work indicated that megalin, part of the apical receptor-mediated endocytic apparatus, may play a role in the PTH-induced removal of NaPi-IIa. Here we examined in rats the time-dependent internalization route of NaPi-IIa after acute PTH application using immunohistochemistry and markers of several endocytic compartments. NaPi-IIa removal from the BBM was detectable as early as 5 min after PTH injection. After 10-15 min, NaPi-IIa was localized in subapical compartments positive for clathrin. Shortly thereafter, NaPi-IIa appeared in endosomes stained for EEA1 (early endosomal antigen 1). After 45-60 min, NaPi-IIa was found in late endosomes/lysosomes marked with lgp120. In contrast, no change in the subcellular localization of megalin and the Na(+)/H(+) exchanger NHE3 was detected up to 60 min after PTH injection. To further characterize the internalization route, insulin, as a marker for receptor-mediated endocytosis, and horseradish peroxidase (HRP) and fluorescein isothiocyanate (FITC)-dextran (10 kDa), as markers for fluid-phase mediated endocytosis, were used. NaPi-IIa colocalized with insulin 5-30 min after PTH injection but did not overlap with HRP or FITC-dextran. These results demonstrate a distinct internalization route of NaPi-IIa in response to acute PTH application that may involve the receptor-mediated endocytic pathway including clathrin-coated vesicles and EEA1-positive early endosomes, and routes NaPi-IIa to lysosomes for degradation.


Subject(s)
Endocytosis/drug effects , Parathyroid Hormone/pharmacology , Sodium-Phosphate Cotransporter Proteins, Type IIa/metabolism , Animals , Clathrin/analysis , Coated Vesicles/chemistry , Coated Vesicles/physiology , Endocytosis/physiology , Endosomes/chemistry , Endosomes/physiology , Fluorescent Antibody Technique , Immunohistochemistry/methods , Insulin/analysis , Insulin/physiology , Kidney/chemistry , Kidney/physiology , Low Density Lipoprotein Receptor-Related Protein-2/analysis , Low Density Lipoprotein Receptor-Related Protein-2/physiology , Lysosomes/chemistry , Lysosomes/physiology , Male , Parathyroid Hormone/administration & dosage , Rats , Rats, Wistar , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/analysis , Sodium-Hydrogen Exchangers/physiology
10.
J Physiol ; 567(Pt 1): 21-6, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-15890704

ABSTRACT

Regulation of renal proximal tubular reabsorption of phosphate (Pi) is one of the critical steps in Pi homeostasis. Experimental evidence suggests that this regulation is achieved mainly by controlling the apical expression of the Na+-dependent Pi cotransporter type IIa (NaPi-IIa) in proximal tubules. Only recently have we started to obtain information regarding the molecular mechanisms that control the apical expression of NaPi-IIa. The first critical observation was the finding that truncation of only its last three amino acid residues has a strong effect on apical expression. A second major finding was the observation that the last intracellular loop of NaPi-IIa contains sequence information that confers parathyroid hormone (PTH) sensitivity. The use of the above domains of the cotransporter in yeast two-hybrid (Y2H) screening allowed the identification of proteins interacting with NaPi-IIa. Biochemical and morphological, as well as functional, analyses have allowed us to obtain insights into the physiological roles of such interactions, although our present knowledge is still far from complete.


Subject(s)
Kidney Tubules, Proximal/metabolism , Phosphates/metabolism , Symporters/metabolism , Animals , Humans , Sodium/metabolism , Sodium-Phosphate Cotransporter Proteins , Sodium-Phosphate Cotransporter Proteins, Type IIa
11.
J Membr Biol ; 206(3): 227-38, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16456717

ABSTRACT

The type IIa Na(+)/P(i), cotransporter (NaPi-IIa) mediates electrogenic transport of three Na(+) and one divalent P(i) ion (and one net positive charge) across the cell membrane. Sequence comparison of electrogenic NaPi-IIa and IIb isoforms with the electroneutral NaPi-IIc isoform pointed to the third transmembrane domain (TMD-3) as a possibly significant determinant of substrate binding. To elucidate the role of TMD-3 in the topology and mechanism underlying NaPi-IIa function we subjected it to cysteine scanning mutagenesis. The constructs were expressed in Xenopus oocytes and P(i) transport kinetics were assayed by electrophysiology and radiotracer uptake. Cys substitution resulted in only marginally altered kinetics of P(i) transport in those mutants providing sufficient current for analysis. Only one site, at the extracellular end of TMD-3, appeared to be accessible to methanethiosulfonate reagents. However, additional mutations carried out at D224 (replaced by E, G or N) and N227 (replaced by D or Q) resulted in markedly altered voltage and substrate dependencies of the P(i)-dependent currents. Replacing Asp-224 (highly conserved in electrogenic a and b isoforms) with Gly (the residue found in the electroneutral c isoform) resulted in a mutant that mediated electroneutral Na(+)-dependent P(i) transport. Since electrogenic NaPi-II transports 3 Na(+)/transport cycle, whereas electroneutral NaPi-IIc only transports 2, we speculate that this loss of electrogenicity might result from the loss of one of the three Na(+) binding sites in NaPi-IIa.


Subject(s)
Cell Membrane/chemistry , Cell Membrane/metabolism , Ion Channel Gating/physiology , Membrane Potentials/physiology , Oocytes/physiology , Sodium-Phosphate Cotransporter Proteins, Type IIa/chemistry , Sodium-Phosphate Cotransporter Proteins, Type IIa/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Cells, Cultured , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Structure-Activity Relationship , Xenopus laevis
12.
Am J Physiol Gastrointest Liver Physiol ; 287(1): G143-50, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15044175

ABSTRACT

Serum and glucocorticoid-inducible kinase 1 (SGK1) is highly expressed in enterocytes. The significance of the kinase in regulation of intestinal function has, however, remained elusive. In Xenopus laevis oocytes, SGK1 stimulates the epithelial Na(+) channel by phosphorylating the ubiquitin ligase Nedd4-2, which regulates channels by ubiquitination leading to subsequent degradation of the channel protein. Thus the present study has been performed to explore whether SGK1 regulates transport systems expressed in intestinal epithelial cells, specifically type IIb sodium-phosphate (Na(+)-P(i)) cotransporter (NaPi IIb). Immunohistochemistry in human small intestine revealed SGK1 colocalization with Nedd4-2 in villus enterocytes. For functional analysis cRNA encoding NaPi IIb, the SGK isoforms and/or the Nedd4-2 were injected into X. laevis oocytes, and transport activity was quantified as the substrate-induced current (I(P)). Exposure to 3 mM phosphate induces an I(P) in NaPi IIb-expressing oocytes. Coinjection of Nedd4-2, but not the catalytically inactive mutant (C938S)Nedd4-2, significantly downregulates I(P), whereas the coinjection of (S422D)SGK1 markedly stimulates I(P) and even fully reverses the effect of Nedd4-2 on I(P). The effect of (S422D)SGK1 on NaPi IIb is mimicked by wild-type SGK3 but not by wild-type SGK2, constitutively active (T308D,S473D)PKB, or inactive (K127N)SGK1. Moreover, (S422D)SGK1 and SGK3 phosphorylate Nedd4-2. In conclusion, SGK1 stimulates the NaPi IIb, at least in part, by phosphorylating and thereby inhibiting Nedd4-2 binding to its target. Thus the present study reveals a novel signaling pathway in the regulation of intestinal phosphate transport, which may be important for regulation of phosphate balance.


Subject(s)
Carrier Proteins/physiology , Ileum/metabolism , Nuclear Proteins , Protein Serine-Threonine Kinases/physiology , Symporters/metabolism , Animals , Cells, Cultured , Enterocytes/metabolism , Humans , Ileum/cytology , Immediate-Early Proteins , Membrane Proteins , Mice , Patch-Clamp Techniques , Phosphorylation , Sodium-Phosphate Cotransporter Proteins , Sodium-Phosphate Cotransporter Proteins, Type IIb , Xenopus laevis
13.
Pflugers Arch ; 448(4): 402-10, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15007650

ABSTRACT

Sodium-dependent phosphate cotransport in renal proximal tubules (PTs) is heterogeneous with respect to proximal tubular segmentation (S1 vs. S3) and nephron generation (superficial vs. juxtamedullary). In the present study, S1 and S3 segments of superficial and juxtamedullary nephrons were laser-microdissected and mRNA and protein expression of the Na/Pi-cotransporters NaPi-IIa and NaPi-IIc and the PDZ proteins NHERF-1 and PDZK1 determined. Expression of NaPi-IIa mRNA decreased axially in juxtamedullary nephrons. There was no effect of dietary Pi content on NaPi-lla mRNA expression in any proximal tubular segment. The abundance of the NaPi-IIa cotransporter in the brush-border membrane showed inter- and intranephron heterogeneity and increased in response to a low-Pi diet (5 days), suggesting that up-regulation of NaPi-lla occurs via post-transcriptional mechanisms. In contrast, NaPi-IIc mRNA and protein was up-regulated by the low-Pi diet in all nephron generations analysed. NHERF-1 and PDZK1, at both mRNA and protein levels, were distributed evenly along the PTs and did not change after a low-Pi diet.


Subject(s)
Kidney Tubules, Proximal/metabolism , Phosphates/metabolism , Symporters/genetics , Symporters/metabolism , Animals , Blotting, Western , Gene Expression , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred Strains , Phosphoproteins/genetics , Phosphoproteins/metabolism , RNA, Messenger/analysis , Sodium-Hydrogen Exchangers , Sodium-Phosphate Cotransporter Proteins , Sodium-Phosphate Cotransporter Proteins, Type IIa , Transcription, Genetic/physiology
14.
J Membr Biol ; 194(2): 91-6, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-14502433

ABSTRACT

In mammals the type IIb Na/P(i)-cotransporter is expressed in various tissues such as intestine, brain, lung and testis. The type IIb cotransporter shows 51% homology with the renal type IIa Na/P(i)-cotransporter, for which a detailed model of the secondary structure has emerged based on recent structure/function studies. To make the type IIb Na/P(i)-cotransporter available for future structural studies, we have expressed this cotransporter in Sf9 cells. Sf9 cells were infected with recombinant baculovirus containing 6His NaPi-IIb. Infected cells expressed a polypeptide of approximately 90 kDa, corresponding to a partially glycosylated form of the type IIb cotransporter. Transport studies demonstrated that the type IIb protein expressed in Sf9 cells mediates transport of phosphate in a Na-dependent manner with similar kinetic characteristics (apparent K(m)s for sodium and phosphate and pH dependence) as previously described. Solubilization experiments demonstrated that, in contrast to the type IIa cotransporter, the type IIb can be solubilized by nonionic detergents and that solubilized type IIb Na/P(i)-cotransporter can be purified by Ni-NTA chromatography.


Subject(s)
Cell Membrane/metabolism , Phosphates/pharmacokinetics , Sodium/metabolism , Spodoptera/chemistry , Spodoptera/enzymology , Symporters/chemistry , Symporters/metabolism , Animals , Cell Line , Gene Expression Regulation, Enzymologic , Hydrogen-Ion Concentration , Molecular Weight , Protein Biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sodium-Phosphate Cotransporter Proteins
15.
J Membr Biol ; 188(3): 227-36, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12181613

ABSTRACT

Analysis of rat and mouse proximal tubular brush-border membrane expression of the type IIa Na/P(i)-cotransporter provides evidence for its cleavage in the large extracellular loop (ECL-2). To study functional properties and membrane distribution of this split NaP(i)-IIa transporter we followed two strategies. In one strategy we expressed the transporter as two complementary parts (p40 and p45) in Xenopus laevis oocytes and as another strategy we cleaved the WT protein with trypsin. Both strategies resulted in a split NaP(i)-IIa protein located in the plasma membrane. The two domains were tied together by a disulfide bridge, most likely involving the cysteines 306 and 334. Surface expression of the NaP(i)-IIa fragments was dependent on the presence of both domains. If both domains were coexpressed, the transporter was functional and transport characteristics were identical to those of the WT-NaP(i)-IIa protein. Corresponding to this, the transporter cleaved by trypsin also retains its transport capacity. These data indicate that cleavage of the type IIa Na/P(i)-cotransporter at ECL-2 is compatible with its cotransport function.


Subject(s)
Gene Expression Regulation , Oocytes/cytology , Oocytes/physiology , Symporters/genetics , Symporters/metabolism , Trypsin/pharmacology , Amino Acid Sequence , Animals , Cell Line , Hydrogen-Ion Concentration , Membrane Potentials/physiology , Molecular Sequence Data , Oocytes/drug effects , Sodium-Phosphate Cotransporter Proteins , Sodium-Phosphate Cotransporter Proteins, Type II , Symporters/drug effects , Trypsin/genetics , Xenopus laevis
16.
J Membr Biol ; 187(2): 85-96, 2002 May 15.
Article in English | MEDLINE | ID: mdl-12029367

ABSTRACT

The effects of the arginine-modifying reagent phenylglyoxal on the kinetics of the type IIa Na + /Pi cotransporter expressed in Xenopus, oocytes were studied by means of 32Pi uptake and electrophysiology. Phenylglyoxal incubation induced up to 60% loss of cotransport function but only marginally altered the Na+-leak. Substrate activation and pH dependency remained essentially unaltered, whereas the voltage dependency of Pi-induced change in electrogenic response was significantly reduced. Presteady-state charge movements were suppressed and the equilibrium charge distribution was shifted slightly towards hyperpolarizing potentials. Charge movements in the absence of external Na+ were also suppressed, which indicated that the empty-carrier kinetics were modified. These effects were incorporated into an ordered alternating access model for NaPi-IIa, whereby the arginine modification by phenylglyoxal was modeled as altered apparent electrical distances moved by mobile charges, together with a slower rate of translocation of the electroneutral, fully loaded carrier.


Subject(s)
Arginine/antagonists & inhibitors , Membrane Potentials/drug effects , Phenylglyoxal/pharmacology , Symporters/genetics , Symporters/metabolism , Animals , Arginine/genetics , Arginine/metabolism , Computer Simulation , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/physiology , Ion Channel Gating/genetics , Ion Channel Gating/physiology , Membrane Potentials/physiology , Models, Biological , Oocytes/physiology , Patch-Clamp Techniques/methods , Phosphorus/metabolism , Reproducibility of Results , Sensitivity and Specificity , Sodium-Phosphate Cotransporter Proteins , Xenopus laevis
17.
Pflugers Arch ; 443(2): 306-13, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11713658

ABSTRACT

Inhibition of proximal tubular phosphate (Pi) reabsorption involves, as far as we know, brush border membrane retrieval of the type IIa Na/Pi-cotransporter. The aim of the present study was to analyze whether intracellular cGMP-mediated regulation of Pi reabsorption also involves retrieval of the type IIa Na/Pi-cotransporter, as previously shown for cAMP. Atrial natriuretic peptide (ANP) and nitric oxide (NO) were used to stimulate guanylate cyclase. In vivo perfusion of mice kidneys with either ANP or NO donors resulted in a downregulation of type IIa Na/Pi-cotransporters on the brush border membranes of proximal tubules. These effects were mimicked by activation of protein kinase G with 8Br-cGMP. In in-vitro-perfused mice proximal tubules, ANP was effective when added either to the apical or basolateral perfusate, suggesting the presence of receptors on both membrane sites. The effects of ANP and NO were blocked by the protein kinase G inhibitor LY 83553. Parallel experiments in OK cells, a renal proximal tubule model, provided similar information. Our findings document that cGMP-mediated regulation (ANP and NO) of type IIa Na/Pi-cotransporters also takes place via internalization of the transporter protein.


Subject(s)
Cyclic GMP/analogs & derivatives , Cyclic GMP/physiology , Kidney/metabolism , Symporters/metabolism , Animals , Atrial Natriuretic Factor/pharmacology , Cell Line , Cyclic GMP/pharmacology , Female , In Vitro Techniques , Kidney/cytology , Kidney Tubules, Proximal/metabolism , Mice , Nitric Oxide/pharmacology , Opossums , Perfusion , Sodium-Phosphate Cotransporter Proteins , Sodium-Phosphate Cotransporter Proteins, Type IIa , Tissue Distribution/drug effects
19.
Pflugers Arch ; 442(5): 782-90, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11512035

ABSTRACT

Type IIa and IIb Na+/Pi-cotransporters have different patterns of expression in vivo: IIa is expressed in apical membranes of renal proximal tubules, and IIb in intestinal and lung epithelia. They are found in different subcellular locations when transfected in epithelial cells: IIa is apically expressed in renal proximal cells (OK), but mostly intracellularly in intestinal cells (CaCo2); IIb is apical in both cell types. To identify the domains responsible for the different expression of both cotransporters (in CaCo2), as well as those responsible for the apical expression of IIa (in OK), mutated cotransporters were fused to the Enhanced Green Fluorescent Protein (EGFP), and their expression analyzed by confocal microscopy. We conclude that the apical expression information for CaCo2 is contained within the C-terminal tail of IIb, but is not contained within IIa. From analysis of mutated IIa cotransporters we identified residues, within the C-terminal tail, involved in the apical expression of these cotransporters in OK cells: internal PR-residues and terminal TRL-residues. These signals are functional in OK but not in CaCo2-cells, supporting the concept that polarized targeting can be protein and cell specific.


Subject(s)
Cell Polarity , Kidney/metabolism , Symporters/genetics , Amino Acid Motifs , Animals , Caco-2 Cells , Cell Line , Green Fluorescent Proteins , Humans , Luminescent Proteins/metabolism , Mice , Microscopy, Confocal , Models, Molecular , Opossums , Protein Isoforms , Recombinant Fusion Proteins/metabolism , Sodium-Phosphate Cotransporter Proteins , Sodium-Phosphate Cotransporter Proteins, Type IIa , Symporters/chemistry , Symporters/metabolism , Transfection
20.
Curr Opin Nephrol Hypertens ; 10(5): 555-61, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11496046

ABSTRACT

The type IIa sodium/inorganic phosphate co-transporter is the rate-limiting inorganic phosphate transport pathway in renal brush-border membranes, and is thus a key player in overall inorganic phosphate homeostasis. Its regulation is mostly associated with membrane retrieval/reinsertion (traffic) of the transport protein. This membrane traffic is controlled by specific 'motifs' at the level of the transporter protein and probably involves interacting proteins (e.g. for scaffolding, regulation or sorting). The intracellular signaling mechanisms (e.g. the involvement of kinases) and the involvement of the cytoskeleton are not yet understood. Hereditary alterations in renal inorganic phosphate handling can be associated with factors controlling the expression of the brush-border type IIa sodium/inorganic phosphate co-transporter.


Subject(s)
Kidney/metabolism , Phosphates/metabolism , Symporters/physiology , Absorption/physiology , Amino Acid Motifs/physiology , Animals , Humans , Sodium-Phosphate Cotransporter Proteins , Sodium-Phosphate Cotransporter Proteins, Type IIa , Symporters/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...