Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Cell Biochem Funct ; 37(6): 400-407, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31328801

ABSTRACT

There are two types of bisphosphonates (BPs), nitrogen-containing (N-BPs) and those free from nitrogen (non-N-BPs). Although N-BPs show greater inhibition of bone resorption than non-N-BPs, their effects are likely accompanied with inflammation, which non-N-BPs mitigate. We examined the competitive effects of zoledronate (ZOL), an N-BP, and etidronate (ETI), a non-N-BP, in osteoblasts. ZOL, but not ETI, markedly reduced alkaline phosphatase activity and cell viability in osteoblastic MC3T3-E1 and Saos2 cells, while that inhibition was relieved by simultaneous administration of ETI, possibly because of competition with ZOL for cellular uptake. However, phosphonoformate, an inhibitor of the phosphonate transporters SLC20A and SLC34A, did not mitigate the reducing effects of ZOL, suggesting that those transporters are not involved in BP uptake in osteoblastic cells. Additionally, ZOL reduced fibroblastic NIH3T3 and C3H10T1/2 cell viability, which was relieved by administration of both ETI and phosphonoformate. Transporter gene expression levels were significantly lower in osteoblasts as compared with fibroblasts, which may account for the distinct effects of phosphonoformate with different cell types. Together, our results suggest existence of a common uptake route of N-BPs and non-N-BPs into osteoblastic cells that is unrelated to the SLC20A and SLC34A families. SIGNIFICANCE OF THE STUDY: N-BP ZOL was shown to suppress differentiation and viability of osteoblasts. ZOL-induced cell viability suppression was also observed in fibroblasts, which was markedly relieved by addition of the non-N-BP ETI. Additionally, mitigation of the effects of ZOL was achieved with phosphonoformate, a sodium-phosphate cotransporter inhibitor, in fibroblastic cells but not osteoblasts. Expression levels of SLC20A and SLC34A family genes were significantly lower in osteoblasts as compared with fibroblasts. These observations suggest that incorporation of N-BPs and non-N-BPs in osteoblasts is mediated via common transporters that appear to be distinct from SLC20A and 34A, which operate in fibroblasts.


Subject(s)
Diphosphonates/pharmacology , Osteoblasts/drug effects , Sodium-Phosphate Cotransporter Proteins/antagonists & inhibitors , 3T3 Cells , Animals , Cell Differentiation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Mice , Osteoblasts/cytology , Osteoblasts/metabolism , Structure-Activity Relationship
2.
Biopharm Drug Dispos ; 39(2): 83-87, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29214648

ABSTRACT

We previously reported the contribution of sodium-phosphate cotransporter to the tubular reabsorption of lithium in rats. In the present study, the dose dependency of the renal handling of lithium was examined in rats. When lithium chloride at 1.25 mg/kg, 2.5 mg/kg and 25 mg/kg was intravenously injected as a bolus, the areas under the plasma concentration-time curve of lithium until 60 minutes were calculated to be 6.23 mEq·min/l, 8.77 mEq·min/l and 64.6 mEq·min/l, respectively. The renal clearance of lithium and its fractional excretion increased with increments in the dose administered. The renal clearance of lithium strongly correlated with the urinary excretion rate of phosphate in the 1.25 mg/kg group (r = 0.840) and 2.5 mg/kg group (r = 0.773), whereas this correlation was weak in the 25 mg/kg group (r = 0.306). The infusion of foscarnet, a typical inhibitor of sodium-phosphate cotransporter, decreased the fractional reabsorption of lithium in rats administered lithium chloride at 2.5 mg/kg, but did not affect it in rats administered 25 mg/kg. These results demonstrate the nonlinearity of the renal excretion of lithium in rats, with the saturation of lithium reabsorption by the sodium-phosphate cotransporter potentially being involved.


Subject(s)
Kidney Tubules/metabolism , Lithium/pharmacokinetics , Renal Reabsorption/drug effects , Sodium-Phosphate Cotransporter Proteins/metabolism , Animals , Dose-Response Relationship, Drug , Foscarnet/pharmacology , Kidney Tubules/drug effects , Lithium/blood , Lithium/urine , Male , Phosphates/urine , Rats , Sodium-Phosphate Cotransporter Proteins/antagonists & inhibitors
3.
Drugs R D ; 13(3): 165-73, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24000048

ABSTRACT

Hyperphosphatemia in chronic kidney disease (CKD) has been associated with elevated cardiovascular morbidity and mortality. Serum phosphate control remains a cornerstone of the clinical management of patients with CKD, in order to both attenuate the progression of secondary hyperparathyroidism or bone disease and (possibly) reduce the risk of vascular calcification. Despite technical improvements in dialysis and the use of dietary restrictions, drug therapy is often required to control phosphate levels in patients with end-stage renal disease (ESRD). Currently available medications for hyperphosphatemia in ESRD are very expensive and not always well tolerated. The discovery and development of new drugs in this indication is therefore a priority for both medical and health-economic reasons. Nicotinamide (an amide derivative of the water-soluble vitamin B3) is a potentially interesting alternative to phosphate binders. In vitro and in vivo data show that nicotinamide reduces hyperphosphatemia by inhibiting sodium-dependent phosphate co-transport in the renal proximal tubule and in the intestine. Accordingly, targeting the sodium-dependent phosphate co-transporter 2b by using nicotinamide as an alternative or adjunct to classical phosphate binders may be a therapeutic option for modulating serum phosphate in CKD. Several recent clinical studies have explored the potential value of nicotinamide in phosphate control (as well as its effects on lipid levels) in dialysis patients. However, we consider that more data on pharmacodynamics, pharmacokinetics and safety are needed before this compound can be recommended as a treatment for hyperphosphatemia in ESRD patients.


Subject(s)
Hyperphosphatemia/drug therapy , Niacinamide/therapeutic use , Renal Dialysis , Humans , Hyperphosphatemia/blood , Hyperphosphatemia/etiology , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/therapy , Niacinamide/administration & dosage , Niacinamide/pharmacokinetics , Phosphates/blood , Renal Dialysis/adverse effects , Sodium-Phosphate Cotransporter Proteins/antagonists & inhibitors , Tissue Distribution
4.
Biochim Biophys Acta ; 1830(3): 2683-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23671929

ABSTRACT

BACKGROUND: Proliferation of Leishmania infantum depends on exogenous inorganic phosphate (P(i)) but little is known about energy metabolism and transport of P(i) across the plasma membrane in Leishmania sp. METHODS: We investigated the kinetics of 32P(i) transport, the influence of H+ and K+ ionophores and inhibitors, and expression of the genes for the Na+:P(i) and H+:P(i) cotransporters. RESULTS: The proton ionophore FCCP, bafilomycin A1 (vacuolar ATPase inhibitor), nigericin (K+ ionophore) and SCH28080 (an inhibitor of H+, K(+)-ATPase) all inhibited the transport of P(i). This transport showed Michaelis-Menten kinetics with K0.5 and V(max) values of 0.016 +/- 0.002 mM and 564.9 +/- 18.06 pmol x h(-1) x 10(-7) cells, respectively. These values classify the P(i) transporter of L. infantum among the high-affinity transporters, a group that includes Pho84 of Saccharomyces cerevisiae. Two sequences were identified in the L. infantum genome that code for phosphate transporters. However, transcription of the PHO84 transporter was 10-fold higher than the PHO89 transporter in this parasite. Accordingly, P(i) transport and LiPho84 gene expression were modulated by environmental P(i) variations. CONCLUSIONS: These findings confirm the presence of a P(i) transporter in L. infantum, similar to PHO84 in S. cerevisiae, that contributes to the acquisition of inorganic phosphate and could be involved in growth and survival of the promastigote forms of L. infantum. GENERAL SIGNIFICANCE: This work provides the first description of a PHO84-like P(i) transporter in a Trypanosomatide parasite of the genus Leishmania, responsible for many infections worldwide.


Subject(s)
Leishmania infantum/enzymology , Phosphates/metabolism , Protozoan Proteins/metabolism , Amino Acid Sequence , Biological Transport , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Culture Media , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Imidazoles/pharmacology , Kinetics , Leishmania infantum/genetics , Macrolides/pharmacology , Molecular Sequence Data , Nigericin/pharmacology , Phosphates/pharmacology , Phosphorus Radioisotopes , Phylogeny , Proton Ionophores/pharmacology , Proton-Phosphate Symporters/antagonists & inhibitors , Proton-Phosphate Symporters/genetics , Proton-Phosphate Symporters/metabolism , Protozoan Proteins/genetics , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sodium-Phosphate Cotransporter Proteins/antagonists & inhibitors , Sodium-Phosphate Cotransporter Proteins/metabolism , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/metabolism , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Vacuolar Proton-Translocating ATPases/metabolism
5.
Drugs Today (Barc) ; 47(12): 923-34, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22348917

ABSTRACT

Walking the streets in Lisbon is not a matter of inertia, at least when going uphill through the small streets crowding the slopes around the Alfama and Bairro Alto neighborhoods. However, when treating diabetes, clinical inertia seems to be the rule, despite the availability of effective therapies and recommendations for early insulin replacement to improve glucose control and prevent diabetes complications. This was further confirmed in the SOLVE study presented during this year's European Association for the Study of Diabetes (EASD) meeting (Khunti, K. et al., Abst 377), highlighting the importance of understanding healthcare habits to implement policies aimed at appropriate treatment intensification and even initiation to achieve earlier glycemic control of type 2 diabetes. However, preventing type 2 diabetes is crucial, in which regard physical activity has been repeatedly reported to reduce the risk of acquiring type 2 diabetes, and does so independently of its impact on general and abdominal adiposity (Ekelund, U. et al., Abst 225), resulting in a clearly cost-effective option to avoid the disease and the need for treatments. Although physical activity improves fitness but not glycemic control in type 1 diabetes (Valletta, J.J. et al., Abst 602), this is feasible through educational campaigns aimed at improving eating habits, routine physical exercise and healthy lifestyles, which, as demonstrated among students in Mexico, reduced the incidence of obesity and overweight (Martínez, M.E., Abst 858). Nevertheless, type 2 diabetes is a reality that is actually on the rise and requires treatments aimed at maintaining glycemic control, thus avoiding diabetic complications, while minimizing the risk of hypoglycemia and its consequences. Reducing hemoglobin A1c levels below 7%, as recommended by most guidelines for the treatment of diabetes, was confirmed to be associated with a reduced risk of death and diabetes-related morbidity in a population study in patients with type 2 diabetes (Skriver, M.V. et al., Abst 54). Hence, treatments are critical for achieving glycemic goals and preventing undue deaths. A wide range of therapies are currently available, but new drugs are under research that may potentially improve outcomes with a lower risk for adverse events, or may offer efficacy in patients currently not at goal because of insufficient efficacy of the drugs they are receiving, or because the doses they would require would cause excessive toxicity or an undue risk for hypoglycemia. New findings with currently available drugs and drugs in research that were reported during this year's EASD meeting in the beautiful city of Lisbon are summarized in the following report.


Subject(s)
Diabetes Mellitus/drug therapy , Diabetes Mellitus/therapy , Hypoglycemic Agents/therapeutic use , Biguanides/therapeutic use , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Enzyme Activators/therapeutic use , Glucokinase/metabolism , Humans , Incretins/therapeutic use , Insulin/administration & dosage , Insulin/analogs & derivatives , Insulin/therapeutic use , Islets of Langerhans Transplantation , Potassium Channel Blockers/pharmacology , Potassium Channel Blockers/therapeutic use , Sodium-Phosphate Cotransporter Proteins/antagonists & inhibitors , Sulfonylurea Compounds/therapeutic use , Thiazolidinediones/therapeutic use
7.
Clin Calcium ; 19(2): 242-7, 2009 Feb.
Article in Japanese | MEDLINE | ID: mdl-19182366

ABSTRACT

In 2003, the NKF K/DOQI released new clinical practice guidelines for bone metabolism and disease in chronic kidney disease, respectively. Target level of phosphate and Ca x Pi products are set at relative low levels in the new guidelines. Prescription of a new phosphate binder, severamer hydrochloride, a widely recommended in this quideline. Other approaches that could prove fruitful include inhibition of intestinal and renal phosphate transporters. Inhibition of the sodium-dependent phosphate transporter by a small molecule would be a desirable method to control serum phosphate levels in patients with chronic renal disease or undergoning dialysis. In this review the progress in the identification of such an agent is discussed.


Subject(s)
Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Drug Design , Foscarnet/therapeutic use , Hyperphosphatemia/drug therapy , Phloretin/analogs & derivatives , Sodium-Phosphate Cotransporter Proteins/antagonists & inhibitors , Animals , Chronic Disease , Foscarnet/pharmacology , Humans , Hyperphosphatemia/etiology , Kidney Diseases/complications , Phloretin/pharmacology , Phloretin/therapeutic use , Practice Guidelines as Topic
8.
Toxicol Appl Pharmacol ; 232(1): 125-34, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18586044

ABSTRACT

Inorganic arsenate (As(V)) is a common contaminant of underground water. Following oral exposure, it is assumed that As(V) is distributed and crosses cell membranes through inorganic phosphate (Pi) transporters. We have tested this hypothesis by studying the inhibition of rat Na/Pi cotransporters by As(V) in Xenopus laevis oocytes and in several rat tissues. The ubiquitously expressed type III Pi transporters (PiT-1 and PiT-2) showed a low affinity for As(V) (K(i) approximately 3.8 mM), similar to the Pi transport system in aortic vascular smooth muscle cells (K(i) 1.5 mM). The type II renal isoforms, NaPi-IIa and NaPi-IIc, were also poorly inhibited by As(V) (K(i) approximately 1 mM), similar to the Pi transport from kidney cortex brush-border membrane (BBM) vesicles. Conversely, the high-affinity intestinal transporter, NaPi-IIb, was very efficiently inhibited with a K(i) of 51 microM, similar to the Pi transport from intestinal BBM vesicles. Taking into account the 1.1 mM Pi in blood and renal ultrafiltrate, and the nanomolar range of As(V) exposures, we have determined that the contribution by Na/Pi cotransporters to As(V) membrane transport is negligible, given that 10-15 mM As(V) would be necessary in these fluids to be significantly transported. Intestinal transport is an exception, because Pi competition is weak, thereby considering that its concentration in lumen mainly depends on low Pi levels from ingested fresh water, and because As(V) very efficiently inhibits Pi intestinal transport. Our data agree with current toxicokinetic knowledge, and they explain the asymmetric excretion of trivalent and pentavalent arsenic species into bile and urine.


Subject(s)
Arsenates/metabolism , Cell Membrane/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIb/metabolism , Sodium-Phosphate Cotransporter Proteins/metabolism , Water Pollutants, Chemical/metabolism , Animals , Arsenates/toxicity , Cells, Cultured , Cloning, Molecular , Dose-Response Relationship, Drug , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Kidney Tubules, Proximal/metabolism , Kinetics , Myocytes, Smooth Muscle/metabolism , Oocytes , Rats , Sodium-Phosphate Cotransporter Proteins/antagonists & inhibitors , Sodium-Phosphate Cotransporter Proteins/genetics , Sodium-Phosphate Cotransporter Proteins, Type III/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIa/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIb/antagonists & inhibitors , Sodium-Phosphate Cotransporter Proteins, Type IIb/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIc/metabolism , Water Pollutants, Chemical/toxicity , Xenopus laevis
9.
Clin J Am Soc Nephrol ; 3(4): 1131-8, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18385391

ABSTRACT

BACKGROUND AND OBJECTIVES: Niacinamide inhibits intestinal sodium/phosphorus transporters and reduces serum phosphorus in open-label studies. A prospective, randomized, double-blind, placebo-controlled crossover trial was performed for assessment of the safety and efficacy of niacinamide. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: Hemodialysis patients with phosphorus levels > or =5.0 mg/dl were randomly assigned to 8 wk of niacinamide or placebo, titrated from 500 to 1500 mg/d. After a 2-wk washout period, patients switched to 8 wk of the alternative therapy. Vitamin D analogs and calcimimetics were held constant; phosphorus binders were not changed unless safety criteria were met. RESULTS: Thirty-three patients successfully completed the trial. Serum phosphorus fell significantly from 6.26 to 5.47 mg/dl with niacinamide but not with placebo (5.85 to 5.98 mg/dl). A concurrent fall in calcium-phosphorus product was seen with niacinamide, whereas serum calcium, intact parathyroid hormone, uric acid, platelet, triglyceride, LDL, and total cholesterol levels remained stable in both arms. Serum HDL levels rose with niacinamide (50 to 61 mg/dl but not with placebo. Adverse effects were similar between both groups. Among patients who were > or =80% compliant, results were similar, although the decrease in serum phosphorus with niacinamide was more pronounced (6.45 to 5.28 mg/dl) and the increase in HDL approached significance (49 to 58 mg/dl). CONCLUSIONS: In hemodialysis patients, niacinamide effectively reduces serum phosphorus when co-administered with binders and results in a potentially advantageous increase in HDL cholesterol. Further study in larger randomized trials and other chronic kidney disease populations is indicated.


Subject(s)
Hyperphosphatemia/drug therapy , Kidney Diseases/therapy , Niacinamide/therapeutic use , Phosphorus/blood , Renal Dialysis , Vitamin B Complex/therapeutic use , Administration, Oral , Biomarkers/blood , Chelating Agents/therapeutic use , Cross-Over Studies , Double-Blind Method , Down-Regulation , Drug Therapy, Combination , Female , Humans , Hyperphosphatemia/blood , Hyperphosphatemia/etiology , Kidney Diseases/blood , Kidney Diseases/complications , Male , Middle Aged , Niacinamide/administration & dosage , Niacinamide/adverse effects , Prospective Studies , Sodium-Phosphate Cotransporter Proteins/antagonists & inhibitors , Treatment Outcome , Vitamin B Complex/administration & dosage , Vitamin B Complex/adverse effects , Vitamin D/therapeutic use , Washington
10.
J Cell Physiol ; 215(1): 47-54, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17894387

ABSTRACT

In this work, we investigated the effect of inorganic phosphate (Pi) on the differentiation of monocyte/macrophage precursors into an "osteoclastic" phenotype, and we delineated the molecular mechanisms which could be involved in this phenomenon. This was achieved by stimulating human peripheral blood monocytic cells and RAW 264.7 monocyte-macrophage precursor cells to differentiate into osteoclast-like cells in the presence of receptor activator of NF-kappaB ligand (RANKL) and macrophage colony-stimulating factor (M-CSF). RANKL has been previously reported to stimulate the signaling kinases ERK 1/2, p38, Akt, JNK, and the DNA-binding activity of the transcription factors AP-1 and NF-kappaB. Increase in extracellular Pi concentration (1.5-4.5 mM) dose-dependently inhibits both osteoclastic differentiation and bone resorption activity induced by RANKL and M-CSF. Pi was found to specifically inhibit the RANKL-induced JNK and Akt activation, while RANKL-induced p38 and ERK 1/2 phosphorylation were not significantly affected. Moreover, we found that Pi significantly reduced the RANKL-stimulated DNA-binding activity of NF-kappaB. The effects of Pi on osteoclast differentiation and DNA-binding activity of NF-kappaB were prevented by Foscarnet, a sodium-phosphate cotransport inhibitor, suggesting that the effects of Pi occur subsequently to its intake. These results demonstrate that Pi downregulates the differentiation of osteoclasts via a negative feedback exerted on RANK-RANKL signaling.


Subject(s)
Extracellular Space/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , Phosphates/pharmacology , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , Acid Phosphatase/metabolism , Animals , Bone Resorption , Cell Differentiation/drug effects , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Foscarnet/pharmacology , Humans , Isoenzymes/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Osteoclasts/cytology , Osteoclasts/enzymology , Osteogenesis/drug effects , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , RANK Ligand/pharmacology , Sodium-Phosphate Cotransporter Proteins/antagonists & inhibitors , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/metabolism , Tartrate-Resistant Acid Phosphatase , Transcription Factor AP-1/metabolism
11.
Drug Metab Pharmacokinet ; 21(3): 217-21, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16858125

ABSTRACT

Inorganic phosphate in food is absorbed two ways, the transcellular route via the brush border membrane and the paracellular route via tight junctions. NaPi, a sodium-dependent inorganic phosphate transporter, is expressed in rat and human intestine. However, the relative contribution of NaPi to total carrier-mediated transport of physiological concentrations of inorganic phosphate in rat intestine is not clear. Here, we characterized inorganic phosphate transport across the rat small intestine using a voltage-clamp analysis which allowed the diffrentiation of inorganic phosphate permeation through these two (transcellular and paracellular) routes. Results showed that, under a physiologically normal transmucosal electrical potential difference (about 2 mV), permeation of inorganic phosphate by the transcellular route was greater than that by the paracellular route. Further, transport was significantly decreased by the addition to the incubation medium of phosphonoformic acid, a sodium-dependent phosphate transporter inhibitor, and severely inhibited under sodium-free conditions. Similar results were obtained without the voltage-clamp. Together, these results suggest that NaPi-mediated transcellular permeation is the dominant route in the absorption of inorganic phosphate across the small intestine.


Subject(s)
Intestinal Absorption/drug effects , Jejunum/metabolism , Phosphates/pharmacokinetics , Sodium-Phosphate Cotransporter Proteins/physiology , Animals , Cell Membrane Permeability/drug effects , Dose-Response Relationship, Drug , Foscarnet/pharmacology , In Vitro Techniques , Intestinal Absorption/physiology , Jejunum/drug effects , Jejunum/physiology , Male , Membrane Potentials/drug effects , Patch-Clamp Techniques/methods , Phosphates/metabolism , Phosphorus Radioisotopes , Potassium Chloride/pharmacology , Rats , Rats, Wistar , Sodium Chloride/pharmacology , Sodium-Phosphate Cotransporter Proteins/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...