Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Curr Opin Nephrol Hypertens ; 31(5): 486-492, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35894284

ABSTRACT

PURPOSE OF REVIEW: Targeting sodium phosphate cotransporter 2a (Npt2a) offers a novel strategy for treating hyperphosphatemia in chronic kidney disease (CKD). Here we review recent studies on the efficacy of Npt2a inhibition, its plasma phosphate (Pi)-lowering effects, as well as potential "off-target" beneficial effects on cardiovascular consequences. RECENT FINDINGS: Two novel Npt2a-selective inhibitors (PF-06869206 and BAY-767) have been developed. Pharmacological Npt2a inhibition shows a significant phosphaturic effect and consequently lowers plasma Pi and parathyroid hormone (PTH) levels regardless of CKD. However, plasma fibroblast growth factor 23 (FGF23), a master regulator of Pi homeostasis, shows inconsistent responses between these two inhibitors (no effect by PF-06869206 vs. reduction by BAY-767). In addition to the effects on Pi homeostasis, Npt2a inhibition also enhances urinary excretions of Na+, Cl-, and Ca2+, which is recapitulated in animal models with reduced kidney function. The effect of Npt2a inhibition by BAY-767 on vascular calcification has been studied, with positive results showing that oral treatment with BAY-767 (10 mg kg-1) attenuated the increases in plasma Pi and Ca2+ content in the aorta under the setting of vascular calcification induced by a pan-FGF receptor inhibitor. Together, Npt2a inhibition offers a promising therapeutic approach for treating hyperphosphatemia and reducing cardiovascular complications in CKD. SUMMARY: Npt2a inhibition significantly increases urinary Pi excretion and lowers plasma Pi and PTH levels; moreover, it exerts pleiotropic "off-target" effects, providing a novel treatment for hyperphosphatemia and exhibiting beneficial potential for cardiovascular complications in CKD.


Subject(s)
Hyperphosphatemia , Renal Insufficiency, Chronic , Sodium-Phosphate Cotransporter Proteins, Type IIa , Vascular Calcification , Animals , Calcium/metabolism , Fibroblast Growth Factors/metabolism , Humans , Hyperphosphatemia/drug therapy , Hyperphosphatemia/etiology , Parathyroid Hormone/metabolism , Phosphates/metabolism , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/drug therapy , Sodium-Phosphate Cotransporter Proteins, Type IIa/antagonists & inhibitors
2.
J Clin Invest ; 130(12): 6510-6522, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32853180

ABSTRACT

The sodium-phosphate cotransporter NPT2a plays a key role in the reabsorption of filtered phosphate in proximal renal tubules, thereby critically contributing to phosphate homeostasis. Inadequate urinary phosphate excretion can lead to severe hyperphosphatemia as in tumoral calcinosis and chronic kidney disease (CKD). Pharmacological inhibition of NPT2a may therefore represent an attractive approach for treating hyperphosphatemic conditions. The NPT2a-selective small-molecule inhibitor PF-06869206 was previously shown to reduce phosphate uptake in human proximal tubular cells in vitro. Here, we investigated the acute and chronic effects of the inhibitor in rodents and report that administration of PF-06869206 was well tolerated and elicited a dose-dependent increase in fractional phosphate excretion. This phosphaturic effect lowered plasma phosphate levels in WT mice and in rats with CKD due to subtotal nephrectomy. PF-06869206 had no effect on Npt2a-null mice, but promoted phosphate excretion and reduced phosphate levels in normophophatemic mice lacking Npt2c and in hyperphosphatemic mice lacking Fgf23 or Galnt3. In CKD rats, once-daily administration of PF-06869206 for 8 weeks induced an unabated acute phosphaturic and hypophosphatemic effect, but had no statistically significant effect on FGF23 or PTH levels. Selective pharmacological inhibition of NPT2a thus holds promise as a therapeutic option for genetic and acquired hyperphosphatemic disorders.


Subject(s)
Hyperphosphatemia/metabolism , Phosphates/metabolism , Renal Insufficiency, Chronic/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIa , Animals , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Hyperphosphatemia/genetics , Hyperphosphatemia/pathology , Male , Mice , Mice, Knockout , N-Acetylgalactosaminyltransferases/genetics , N-Acetylgalactosaminyltransferases/metabolism , Rats , Rats, Sprague-Dawley , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Sodium-Phosphate Cotransporter Proteins, Type IIa/antagonists & inhibitors , Sodium-Phosphate Cotransporter Proteins, Type IIa/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIa/metabolism , Polypeptide N-acetylgalactosaminyltransferase
3.
J Am Soc Nephrol ; 30(11): 2128-2139, 2019 11.
Article in English | MEDLINE | ID: mdl-31409727

ABSTRACT

BACKGROUND: The kidneys play an important role in phosphate homeostasis. Patients with CKD develop hyperphosphatemia in the later stages of the disease. Currently, treatment options are limited to dietary phosphate restriction and oral phosphate binders. The sodium-phosphate cotransporter Npt2a, which mediates a large proportion of phosphate reabsorption in the kidney, might be a good therapeutic target for new medications for hyperphosphatemia. METHODS: The authors assessed the effects of the first orally bioavailable Npt2a inhibitor (Npt2a-I) PF-06869206 in normal mice and mice that had undergone subtotal nephrectomy (5/6 Nx), a mouse model of CKD. Dose-response relationships of sodium, chloride, potassium, phosphate, and calcium excretion were assessed in response to the Npt2a inhibitor in both groups of mice. Expression and localization of Npt2a/c and levels of plasma phosphate, calcium, parathyroid hormone (PTH) and fibroblast growth factor 23 (FGF-23) were studied up to 24-hours after Npt2a-I treatment. RESULTS: In normal mice, Npt2a inhibition caused a dose-dependent increase in urinary phosphate (ED50 approximately 21 mg/kg), calcium, sodium and chloride excretion. In contrast, urinary potassium excretion, flow rate and urinary pH were not affected dose dependently. Plasma phosphate and PTH significantly decreased after 3 hours, with both returning to near baseline levels after 24 hours. Similar effects were observed in the mouse model of CKD but were reduced in magnitude. CONCLUSIONS: Npt2a inhibition causes a dose-dependent increase in phosphate, sodium and chloride excretion associated with reductions in plasma phosphate and PTH levels in normal mice and in a CKD mouse model.


Subject(s)
Hypophosphatemia, Familial/etiology , Phosphates/blood , Renal Insufficiency, Chronic/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIa/antagonists & inhibitors , Animals , Calcium/urine , Disease Models, Animal , Dose-Response Relationship, Drug , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/blood , Male , Mice , Mice, Inbred C57BL , Parathyroid Hormone/blood
4.
Genomics ; 108(2): 56-63, 2016 08.
Article in English | MEDLINE | ID: mdl-27241791

ABSTRACT

Natural antisense transcripts (NATs) can interfere with the expression of complementary sense transcripts with exquisite specificity. We have previously cloned NATs of Slc34a loci (encoding Na-phosphate transporters) from fish and mouse. Here we report the cloning of a human SLC34A1-related NAT that represents an alternatively spliced PFN3 transcript (Profilin3). The transcript is predominantly expressed in testis. Phylogenetic comparison suggests two distinct mechanisms producing Slc34a-related NATs: Alternative splicing of a transcript from a protein coding downstream gene (Pfn3, human/mouse) and transcription from the bi-directional promoter (Rbpja, zebrafish). Expression analysis suggested independent regulation of the complementary Slc34a mRNAs. Analysis of randomly selected bi-directionally transcribed human/mouse loci revealed limited phylogenetic conservation and independent regulation of NATs. They were reduced on X chromosomes and clustered in regions that escape inactivation. Locus structure and expression pattern suggest a NATs-associated regulatory mechanisms in testis unrelated to the physiological role of the sense transcript encoded protein.


Subject(s)
Alternative Splicing , Cloning, Molecular/methods , Profilins/genetics , Profilins/metabolism , RNA, Antisense/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIa/antagonists & inhibitors , Animals , Databases, Genetic , Evolution, Molecular , Gene Expression Regulation , HEK293 Cells , Humans , Kidney/metabolism , Male , Mice , Phylogeny , Promoter Regions, Genetic , RNA, Antisense/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIa/genetics , Testis/metabolism , Tissue Distribution
5.
Drug Metab Pharmacokinet ; 31(3): 256-9, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27238574

ABSTRACT

Lithium, which is used in the treatment of and prophylaxis for bipolar disease, inhibits glycogen synthase kinase-3ß (GSK3ß) by producing its phosphorylated form (p-GSK3ß). GSK3ß plays a role in apoptosis and some kinds of acute kidney injuries, and the formation of p-GSK3ß is considered to contribute to protection against acute kidney injury. We previously reported that the sodium-phosphate cotransporter NaPi-IIa (SLC34A1) mediated the reabsorption of lithium in the rat kidney. In the present study, the phosphorylation status of GSK3ß in the kidney cortex of rats administered lithium chloride and foscarnet, a typical inhibitor of NaPi-IIa, was examined using Western blotting. Under a 2-h infusion of lithium chloride, the plasma concentration of lithium was 1.06 mEq/l, and its renal clearance was calculated as 1.18 ml/min/kg, which was 29.6% of creatinine clearance. The abundance of p-GSK3ß in the kidney cortex was augmented by the administration of lithium. The simultaneous infusion of foscarnet increased the renal clearance of lithium and its ratio to creatinine clearance as well as the urinary excretion of phosphate. Foscarnet also inhibited the lithium-induced phosphorylation of GSK3ß. These results suggest that the reabsorption of lithium by NaPi-IIa triggers the phosphorylation of GSK3ß in the rat kidney cortex.


Subject(s)
Antimanic Agents/pharmacology , Foscarnet/pharmacology , Glycogen Synthase Kinase 3 beta/metabolism , Kidney Cortex/drug effects , Lithium Chloride/pharmacology , Sodium-Phosphate Cotransporter Proteins, Type IIa/antagonists & inhibitors , Animals , Kidney Cortex/enzymology , Lithium Chloride/antagonists & inhibitors , Male , Phosphorylation/drug effects , Rats, Wistar
6.
Kidney Blood Press Res ; 37(6): 547-56, 2013.
Article in English | MEDLINE | ID: mdl-24356547

ABSTRACT

BACKGROUND/AIMS: The Na(+)-coupled phosphate transporter NaPi-IIa is the main carrier accomplishing renal tubular phosphate reabsorption. It is driven by the electrochemical Na(+) gradient across the apical cell membrane, which is maintained by Na(+) extrusion across the basolateral cell membrane through the Na(+)/K(+) ATPase. The operation of NaPi-IIa thus requires energy in order to avoid cellular Na(+) accumulation and K(+) loss with eventual decrease of cell membrane potential, Cl(-) entry and cell swelling. Upon energy depletion, early inhibition of Na(+)-coupled transport processes may delay cell swelling and thus foster cell survival. Energy depletion is sensed by the AMP-activated protein kinase (AMPK), a serine/threonine kinase stimulating several cellular mechanisms increasing energy production and limiting energy utilization. The present study explored whether AMPK influences the activity of NAPi-IIa. METHODS: cRNA encoding NAPi-IIa was injected into Xenopus oocytes with or without additional expression of wild-type AMPK (AMPK(α1)-HA+AMPK(ß1)-Flag+AMPK(γ1)-HA), of inactive AMPK(αK45R) (AMPK(α1K45R)+AMPK(ß1)-Flag+AMPK(γ1)-HA) or of constitutively active AMPK(γR70Q) (AMPK(α1)-HA+AMPK(ß1)-Flag+AMPKγ1(R70Q)). NaPi-IIa activity was estimated from phosphate-induced current in dual electrode voltage clamp experiments. RESULTS: In NaPi-IIa-expressing, but not in water-injected Xenopus oocytes, the addition of phosphate (1 mM) to the extracellular bath solution generated a current (Ip), which was significantly decreased by coexpression of wild-type AMPK and of AMPK(γR70Q) but not of AMPK(αK45R). The phosphate-induced current in NaPi-IIa- and AMPK-expressing Xenopus ooocytes was significantly increased by AMPK inhibitor Compound C (20 µM). Kinetic analysis revealed that AMPK significantly decreased the maximal transport rate. CONCLUSION: The AMP-activated protein kinase AMPK is a powerful regulator of NaPi-IIa and thus of renal tubular phosphate transport. © 2013 S. Karger AG, Basel.


Subject(s)
AMP-Activated Protein Kinases/physiology , Down-Regulation/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIa/antagonists & inhibitors , AMP-Activated Protein Kinases/genetics , Animals , Catalytic Domain/genetics , Female , Luminescence , Mutation , Oocytes/enzymology , Oocytes/metabolism , Protein Kinase Inhibitors/pharmacology , Sodium-Phosphate Cotransporter Proteins, Type IIa/biosynthesis , Sodium-Phosphate Cotransporter Proteins, Type IIa/genetics , Xenopus laevis
7.
Eur J Pharmacol ; 721(1-3): 332-40, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24056120

ABSTRACT

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.


Subject(s)
Biological Assay/methods , Sodium-Phosphate Cotransporter Proteins, Type IIa/metabolism , Biological Transport/drug effects , Dose-Response Relationship, Drug , Doxycycline/pharmacology , Drug Evaluation, Preclinical , HEK293 Cells , Humans , Phosphates/metabolism , Sesquiterpenes/pharmacology , Sodium/pharmacology , Sodium-Phosphate Cotransporter Proteins, Type IIa/antagonists & inhibitors , Sodium-Phosphate Cotransporter Proteins, Type IIa/genetics , Time Factors , Transfection
8.
Toxicol Sci ; 101(2): 254-62, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17998273

ABSTRACT

It has been suggested that uranium uptake and toxicity could be mediated by endocytosis and/or the type IIa sodium-dependent phosphate cotransporter (NaPi-IIa). The aim of this study was therefore to characterize in vitro the role of these two cellular mechanisms in the uptake and toxicity of low (200-3200 nM) and high (0.5 and 0.8 mM) concentrations of uranium, respectively. At low concentrations, uranium uptake in LLC-PK(1) cells was saturable (V(max) = 3.09 +/- 0.22 ng/mg protein) and characterized by a K(0.5) of 1022 +/- 63 nM and a Hill coefficient of 3.0 +/- 0.4. The potential involvement of endocytosis and NaPi-IIa in the uptake of uranium was assessed by the use of various drugs and culture conditions known to alter their relative activity, and (233)uranium uptake was monitored. Interestingly, the inhibitory effect of colchicine, cytochalasin D, phorbol 12-myristate 13-acetate, and chlorpromazine on endocytosis was highly correlated with their effect on uranium uptake, a relationship that was not true when the NaPi-IIa transport system was studied. Whereas the competitive inhibition of the NaPi-IIa by phosphonoformic acid (PFA) significantly decreased uranium uptake, this effect was not reproduced when NaPi-IIa inhibition was mediated by the replacement of extracellular Na(+) with N-methyl-D-glucamine. Uranium uptake was also not significantly altered when NaPi-IIa expression was stimulated in MDCK cells. More surprisingly, we observed by transmission electron microscopy that uranium cytotoxicity was dependent upon the extent of its intracellular precipitation, but not on its intracellular content, and was suppressed by PFA. In conclusion, our results suggest that low-dose uranium uptake is mainly mediated by absorptive endocytosis, and we propose PFA as a potential uranium chelator.


Subject(s)
Endocytosis , Sodium-Phosphate Cotransporter Proteins, Type IIa/physiology , Uranyl Nitrate , Animals , Biological Transport , Cell Survival/drug effects , Dose-Response Relationship, Drug , Endocytosis/drug effects , Foscarnet/pharmacology , Kinetics , LLC-PK1 Cells , Microscopy, Electron, Transmission , Sodium-Phosphate Cotransporter Proteins, Type IIa/antagonists & inhibitors , Sodium-Phosphate Cotransporter Proteins, Type IIa/metabolism , Swine , Uranyl Nitrate/metabolism , Uranyl Nitrate/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...