Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Kidney Blood Press Res ; 35(5): 365-72, 2012.
Article in English | MEDLINE | ID: mdl-22473073

ABSTRACT

BACKGROUND/AIMS: Gum arabic (GA) is a Ca(2+)-, Mg(2+)- and K(+)-rich dietary fiber used for the treatment of patients with chronic kidney disease in Middle Eastern countries. In healthy mice, GA treatment increases creatinine clearance, renal ADH excretion, as well as intestinal and renal excretion of Mg(2+) and Ca(2+). GA decreases plasma Pi concentration, urinary Pi and Na(+) excretion. The present study explored the effects of GA on renal function in diabetic mice. METHODS: Metabolic cage experiments were performed on Akita mice (akita(+/-)), which spontaneously develop insulin deficiency and thus hyperglycemia. Plasma and urinary concentrations of Na(+), K(+) and Ca(2+) were measured by flame photometry (AFM 5051, Eppendorf, Germany), creatinine by the Jaffé method, phosphate photometrically, urea by an enzymatic method, glucose utilizing a glucometer and an enzymatic kit, aldosterone using an RIA, urinary albumin fluorometrically, and blood pressure by the tail-cuff method. RESULTS: GA (10% in drinking water) significantly increased urinary excretion of Ca(2+) and significantly decreased plasma phosphate and urea concentrations, urinary flow rate, urinary Na(+), phosphate and glucose excretion, blood pressure and proteinuria. CONCLUSIONS: GA treatment decreases blood pressure and proteinuria in diabetic mice and may thus prove beneficial in diabetic nephropathy.


Subject(s)
Diabetic Nephropathies/drug therapy , Gum Arabic/pharmacology , Kidney/drug effects , Kidney/physiology , Animals , Blood Pressure/drug effects , Calcium/blood , Calcium/urine , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/physiopathology , Disease Models, Animal , Gum Arabic/chemistry , Hyperglycemia/drug therapy , Hyperglycemia/metabolism , Hyperglycemia/physiopathology , Hypertension, Renal/drug therapy , Hypertension, Renal/metabolism , Hypertension, Renal/physiopathology , Male , Mice , Mice, Mutant Strains , Potassium/blood , Potassium/urine , Proteinuria/drug therapy , Proteinuria/metabolism , Proteinuria/physiopathology , Sodium/blood , Sodium/urine
2.
Kidney Int ; 80(1): 61-7, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21451460

ABSTRACT

Insulin and growth factors activate the phosphatidylinositide-3-kinase pathway, leading to stimulation of several kinases including serum- and glucocorticoid-inducible kinase isoform SGK3, a transport regulating kinase. Here, we explored the contribution of SGK3 to the regulation of renal tubular phosphate transport. Coexpression of SGK3 and sodium-phosphate cotransporter IIa significantly enhanced the phosphate-induced current in Xenopus oocytes. In sgk3 knockout and wild-type mice on a standard diet, fluid intake, glomerular filtration and urine flow rates, and urinary calcium ion excretion were similar. However, fractional urinary phosphate excretion was slightly but significantly larger in the knockout than in wild-type mice. Plasma calcium ion, phosphate concentration, and plasma parathyroid hormone levels were not significantly different between the two genotypes, but plasma calcitriol and fibroblast growth factor 23 concentrations were significantly lower in the knockout than in wild-type mice. Moreover, bone density was significantly lower in the knockouts than in wild-type mice. Histological analysis of the femur did not show any differences in cortical bone but there was slightly less prominent trabecular bone in sgk3 knockout mice. Thus, SGK3 has a subtle but significant role in the regulation of renal tubular phosphate transport and bone density.


Subject(s)
Bone Density/physiology , Hypophosphatemia, Familial/etiology , Protein Serine-Threonine Kinases/deficiency , Animals , Biological Transport, Active , Bone Density/genetics , Calcium/metabolism , Female , Humans , Hypophosphatemia, Familial/enzymology , Hypophosphatemia, Familial/genetics , In Vitro Techniques , Kidney Tubules/metabolism , Mice , Mice, Knockout , Oocytes/metabolism , Phosphates/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Sodium-Phosphate Cotransporter Proteins, Type IIa/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIa/metabolism , Xenopus
3.
J Am Soc Nephrol ; 22(5): 873-80, 2011 May.
Article in English | MEDLINE | ID: mdl-21493770

ABSTRACT

Insulin and IGF1-dependent signaling activates protein kinase B and serum and glucocorticoid inducible kinase (PKB/SGK), which together phosphorylate and inactivate glycogen synthase kinase GSK3. Because insulin and IGF1 increase renal tubular calcium and phosphorus reabsorption, we examined GSK3 regulation of phosphate transporter activity and determined whether PKB/SGK inactivates GSK3 to enhance renal phosphate and calcium transport. Overexpression of GSK3 and the phosphate transporter NaPi-IIa in Xenopus oocytes decreased electrogenic phosphate transport compared with NaPi-IIa-expressing oocytes. PKB/SGK serine phosphorylation sites in GSK3 were mutated to alanine to create gsk3(KI) mice resistant to PKB/SGK inactivation. Compared with wildtype animals, gsk3(KI) animals exhibited greater urinary phosphate and calcium clearances with higher excretion rates and lower plasma concentrations. Isolated brush border membranes from gsk3(KI) mice showed less sodium-dependent phosphate transport and Na-phosphate co-transporter expression. Parathyroid hormone, 1,25-OH vitamin D levels, and bone mineral density were decreased in gsk3(KI) mice, suggesting a global dysregulation of bone mineral metabolism. Taken together, PKB/SGK phosphorylation of GSK3 increases phosphate transporter activity and reduces renal calcium and phosphate loss.


Subject(s)
Calcium/urine , Glycogen Synthase Kinase 3/physiology , Hypophosphatemia, Familial/etiology , Immediate-Early Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Animals , Bone Density , Calcitriol/blood , Kidney Tubules/metabolism , Mice , Parathyroid Hormone/blood , Phosphates/metabolism , Phosphorylation
4.
Pflugers Arch ; 461(5): 527-36, 2011 May.
Article in English | MEDLINE | ID: mdl-21476133

ABSTRACT

Loss of function mutations of the tumor suppressor gene adenomatous polyposis coli (APC) underly the familial adenomatous polyposis. Mice carrying an inactivating mutation in the apc gene (apc (Min/+)) similarly develop intestinal polyposis. APC is effective at least in part by degrading ß-catenin and lack of APC leads to markedly enhanced cellular ß-catenin levels. ß-Catenin has most recently been shown to upregulate the Na+/K+ ATPase. The present study, thus, explored the possibility that APC could influence intestinal transport. The abundance and localization of ß-catenin were determined utilizing Western blotting and confocal microscopy, the activity of the electrogenic glucose carrier (SGLT1) was estimated from the glucose-induced current in jejunal segments utilizing Ussing chamber experiments and the Na+/H+ exchanger (NHE3) activity from Na+ -dependent re-alkalinization of cytosolic pH (ΔpH(i)) following an ammonium pulse employing BCECF fluorescence. As a result, ß-catenin abundance in intestinal tissue was significantly higher in apc (Min/+) mice than in wild-type mice (apc (+/+)). The ß-catenin protein was localized in the basolateral membrane. Both, the glucose-induced current and ΔpH(i) were significantly higher in apc (Min/+) mice than in apc (+/+) mice. In conclusion, intestinal electrogenic transport of glucose and intestinal Na+/H+ exchanger activity are both significantly enhanced in apc (Min/+) mice, pointing to a role of APC in the regulation of epithelial transport.


Subject(s)
Genes, APC/physiology , Intestinal Mucosa/metabolism , beta Catenin/metabolism , Animals , Biological Transport/genetics , Down-Regulation , Glucose/metabolism , Hydrogen-Ion Concentration , Mice , Sodium-Glucose Transporter 1/metabolism , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/metabolism
5.
Cell Physiol Biochem ; 26(4-5): 641-6, 2010.
Article in English | MEDLINE | ID: mdl-21063101

ABSTRACT

Glycogen synthase kinase 3 GSK3ß participates in a wide variety of functions including regulation of glucose metabolism. It is ubiquitously expressed including epithelial tissues. However, whether GSK3ß participates in the regulation of epithelial transport is not known. The present study thus explored whether GSK3ß influences the Na(+)-coupled transport of glucose. To this end, SGLT1 was expressed in Xenopus oocytes with or without GSK3ß and glucose-induced current (I(g)) determined by dual electrode voltage clamp. In Xenopus oocytes expressing SGLT1 but not in water-injected oocytes glucose induced an inwardly directed I(g), which was significantly enhanced by coexpression of GSK3ß. According to chemiluminescence and confocal microscopy, GSK3ß increased the SGLT1 protein abundance in the oocyte cell membrane. To explore whether GSK3ß sensitivity of SGLT1 participates in the regulation of electrogenic intestinal glucose transport, Ussing chamber experiments were performed in intestinal segments from gene-targeted knockin mice with mutated and thus PKB/SGK-resistant GSK3α,ß (gsk3(KI)), in which the serine of the PKB/SGK phosphorylation site was replaced by alanine, and from wild type mice (gsk3(WT)). The glucose-induced current was significantly larger in gsk3(KI) than in gsk3(WT) mice. The present observations reveal a novel function of GSK3, i.e. the stimulation of Na(+)-coupled glucose transport.


Subject(s)
Glucose/metabolism , Glycogen Synthase Kinase 3/metabolism , 3-Phosphoinositide-Dependent Protein Kinases , Animals , Female , Gene Knock-In Techniques , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Male , Mice , Mutation , Oocytes/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Sodium-Glucose Transporter 1/genetics , Sodium-Glucose Transporter 1/metabolism , Xenopus/growth & development
6.
Am J Physiol Renal Physiol ; 299(5): F1171-7, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20719979

ABSTRACT

Klotho is a membrane protein participating in the inhibitory effect of FGF23 on the formation of 1,25-dihydroxyvitamin-D(3) [1,25(OH)(2)D(3)]. It participates in the regulation of renal tubular phosphate reabsorption and stimulates renal tubular Ca(2+) reabsorption. Klotho hypomorphic mice (klotho(hm)) suffer from severe growth deficit, rapid aging, and early death, events largely reversed by a vitamin D-deficient diet. The present study explored the role of Klotho deficiency in mineral and electrolyte metabolism. To this end, klotho(hm) mice and wild-type mice (klotho(+/+)) were subjected to a normal (D(+)) or vitamin D-deficient (D(-)) diet or to a vitamin D-deficient diet for 4 wk and then to a normal diet (D(-/+)). At the age of 8 wk, body weight was significantly lower in klotho(hm)D(+) mice than in klotho(+/+)D(+) mice, klotho(hm)D(-) mice, and klotho(hm)D(-/+) mice. Plasma concentrations of 1,25(OH)(2)D(3,) adrenocorticotropic hormone (ACTH), antidiuretic hormone (ADH), and aldosterone were significantly higher in klotho(hm)D(+) mice than in klotho(+/+)D(+) mice. Plasma volume was significantly smaller in klotho(hm)D(-/+) mice, and plasma urea, Ca(2+), phosphate and Na(+), but not K(+) concentrations were significantly higher in klotho(hm)D(+) mice than in klotho(+/+)D(+) mice. The differences were partially abrogated by a vitamin D-deficient diet. Moreover, the hyperaldosteronism was partially reversed by Ca(2+)-deficient diet. Ussing chamber experiments revealed a marked increase in amiloride-sensitive current across the colonic epithelium, pointing to enhanced epithelial sodium channel (ENaC) activity. A salt-deficient diet tended to decrease and a salt-rich diet significantly increased the life span of klotho(hm)D(+) mice. In conclusion, the present observation disclose that the excessive formation of 1,25(OH)(2)D(3) in Klotho-deficient mice results in extracellular volume depletion, which significantly contributes to the shortening of life span.


Subject(s)
Glucuronidase/genetics , Glucuronidase/physiology , Hyperaldosteronism/genetics , Adrenocorticotropic Hormone/blood , Aldosterone/blood , Animals , Blood Chemical Analysis , Blood Pressure/physiology , Body Weight/physiology , Calcitriol/metabolism , Diffusion Chambers, Culture , Electrolytes/metabolism , Fibroblast Growth Factor-23 , Hyperaldosteronism/metabolism , Klotho Proteins , Mice , Mice, Knockout , Parathyroid Hormone/blood , Plasma Volume/physiology , Survival , Vasopressins/blood
7.
Cell Physiol Biochem ; 25(6): 695-704, 2010.
Article in English | MEDLINE | ID: mdl-20511715

ABSTRACT

Pharmacological inhibition of phosphoinositol 3 kinase (PI3K) and partial deficiency of phosphoinositide dependent kinase PDK1 have previously been shown to enhance basal gastric acid secretion. PI3K/PDK1 dependent signaling involves activation of protein kinase B/Akt, which may thus be similarly involved in the regulation of gastric acid secretion. To test that hypothesis, gastric acid secretion was determined in isolated glands from gene targeted mice lacking functional Akt2 (akt2(-/-)) or from their wild type littermates (akt2(+/+)). According to BCECF-fluorescence cytosolic pH in isolated gastric glands was similar in akt2(-/-) and akt2(+/+) mice. Na(+)-independent pH recovery (DeltapH/min) following an ammonium pulse, a measure of H(+)/K(+) ATPase activity, was, however, significantly faster in akt2(-/-) than in akt2(+/+) mice. In both genotypes, DeltapH/min was virtually abolished by H(+)/K(+) ATPase inhibitor omeprazole (100 muM). Increase of extracellular K(+) concentrations to 35 mM (replacing Na(+)) increased DeltapH/min to a significantly larger extent in akt2(+/+) than in akt2(-/-) mice and dissipated the differences between the genotypes. Similarly, treatment with 5 muM forskolin enhanced DeltapH/min significantly only in akt2(+/+) mice and abolished the differences between the genotypes. Conversely, protein kinase A inhibitor H89 (50 nM) decreased DeltapH/min to similarly low values in both genotypes. In conclusion, Akt2 suppresses gastric acid secretion and contributes to or even accounts for the inhibition of gastric acid secretion by PI3K.


Subject(s)
Gastric Acid/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cells, Cultured , Cyclic AMP/metabolism , Female , Gastric Mucosa/metabolism , Gene Deletion , Hydrogen-Ion Concentration , KCNQ1 Potassium Channel/metabolism , Male , Mice , Parietal Cells, Gastric/metabolism , Potassium/metabolism , Proto-Oncogene Proteins c-akt/genetics
8.
Cell Physiol Biochem ; 25(6): 715-22, 2010.
Article in English | MEDLINE | ID: mdl-20511717

ABSTRACT

The phosphoinositol 3 kinase (PI3K) and the phosphoinositide dependent kinase (PDK1) stimulate the serum and glucocorticoid inducible kinase (SGK) and protein kinase B (PKB/Akt) isoforms, kinases stimulating a variety of transporters. Most recently, SGK1 was shown to stimulate the peptide transporters PepT1 and PepT2, and to mediate the glucocorticoid stimulation of PepT1. Basal electrogenic intestinal peptide transport was, however, not dependent on the presence of SGK1. The present study explored whether basal electrogenic intestinal peptide transport is dependent on PI3K or PDK1. To this end, peptide transport in intestinal segments was determined utilizing Ussing chamber analysis. Cytosolic pH (pH(i)) was determined by BCECF fluorescence. The luminal addition of 5 mM dipeptide gly-gly induced a current (Ip) across intestinal segments. Ip was significantly decreased in the presence of PI3 kinase inhibitors Wortmannin (1 microM) or LY294002 (50 microM). Exposure of isolated intestinal cells to 5 mM gly-gly was followed by cytosolic acidification (DeltapH(i)), which was significantly blunted by Wortmannin and by LY294002. Both, Ip and DeltapHi were significantly smaller in PDK1 hypomorphic mice (pdk(1flfl)) than in their wild type littermates (pdk1(wt)). In conclusion, PI3K and PDK1 participate in the regulation of basal peptide transport.


Subject(s)
Dipeptides/metabolism , Intestinal Mucosa/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Transport , 3-Phosphoinositide-Dependent Protein Kinases , Animals , Intestines/cytology , Mice
9.
Cell Physiol Biochem ; 25(6): 775-86, 2010.
Article in English | MEDLINE | ID: mdl-20511724

ABSTRACT

BACKGROUND: Mounting evidence suggests that bipolar disorder symptoms could be favorably influenced by modification of glycogen synthase kinase-3 (GSK-3) activity. Specifically, the well known antimanic and mood stabilizing medications lithium, valproate, olanzapine and clozapine have been shown to inhibit GSK-3 activity. GSK-3 is phosphorylated and thus inhibited by protein kinase B (PKB/Akt) and serum and glucocorticoid inducible kinase (SGK) isoforms. The present study explored, whether PKB/SGK-dependent GSK-3 regulation influences the behavior of mice. METHODS: Gene-targeted knockin mice with mutated and thus PKB/SGK-resistant GSK-3alpha, beta (gsk-3(KI)) were compared to corresponding wild type mice (gsk-3(WT)). The mice were analyzed by open-field, light-dark (LD-) box, O-maze, emergence test, object exploration test and forced swimming test (FST). RESULTS: In open-field, LD-box and O-maze, gsk-3(KI) mice displayed a hyperactive and more curious phenotype when compared to wild type mice. Speed and total distance moved as well as rearings were significantly increased in gsk-3(KI) compared to gsk-3(WT) mice. In the O-maze, gsk-3(KI) mice tended to travel a larger distance in the open, unprotected area thangsk-3(WT) mice, and performed significantly more unprotected head dips suggesting decreased anxiety behavior. In the forced swimming test, the immobility time was significantly decreased in gsk-3(KI) mice indicating a phenotype less prone to depression. Moreover, gsk-3(KI)mice were less sensitive to the application of chronic mild stress and showed a decreased HPA axis activity. CONCLUSIONS: The present observations disclose a significant role of PKB/SGK-dependent regulation of GSK-3 in the control of activity, anxiety and proneness to depression. Accordingly, mice expressing SGK/PKB resistant GSK-3 may be a valuable model of hyperactivity and mania.


Subject(s)
Exploratory Behavior , Glycogen Synthase Kinase 3/metabolism , Hyperkinesis/metabolism , Immediate-Early Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Depression/genetics , Depression/metabolism , Female , Gene Expression Regulation , Glycogen Synthase Kinase 3/genetics , Hyperkinesis/genetics , Immediate-Early Proteins/genetics , Male , Mice , Mutation , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics
10.
Pflugers Arch ; 460(3): 667-76, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20490540

ABSTRACT

Annexin A7 is a ubiquitously expressed Ca(2+)- and phospholipid-binding protein. Erythrocytes from mice lacking annexin A7 (anxA7(-/-)) are deformed and relatively resistant to osmotic swelling. In normal erythrocytes, hyperosmotic shock, Cl(-) removal, and energy depletion (glucose removal) trigger PGE(2) formation, which stimulates Ca(2+)-permeable cation channels, increases cytosolic Ca(2+) activity ([Ca(2+)](i)), and thus triggers suicidal death of erythrocytes or eryptosis, characterized by scrambling of the cell membrane with phosphatidylserine exposure at the cell surface. The present experiments explored the influence of annexin A7 deficiency on eryptosis. In erythrocytes from annexin A7-deficient mice (anxA7(-/-)) and wild-type mice (anxA7(+/+)), PGE(2) formation was determined utilizing an immunoassay, ion channel activity by whole-cell patch clamp recording, [Ca(2+)](i) by fluo3 fluorescence, and phosphatidylserine exposure by binding of annexin A5 in fluorescence activated cell sorter (FACS) analysis. Erythrocyte number and hematocrit were significantly smaller in blood from anx7(-/-) than in anx7(+/+) mice. Cl(-)-removal (replacement with gluconate) stimulated PGE(2)-formation, activated cation currents, increased [Ca(2+)](i), and triggered phosphatidylserine exposure, effects significantly more pronounced in anx7(-/-) than in anx7(+/+) erythrocytes. Hyperosmotic shock (addition of 400 mM sucrose) and glucose depletion (removal of glucose) similarly increased cytosolic Ca(2+) activity and triggered phosphatidylserine exposure, effects again significantly more pronounced in anx7(-/-) than in anx7(+/+) erythrocytes. The effects of Cl(-) removal on PGE(2) formation and the cation current, as well as the effect of hypertonic cell shrinkage on [Ca(2+)](i) and cell membrane scrambling, were blunted following inhibition of cyclooxygenase by aspirin or diclofenac. In conclusion, lack of annexin A7 sensitizes the erythrocytes for "proapoptotic" Ca(2+) overload, an effect shortening the life span of the affected erythrocytes and, thus, leading to anemia.


Subject(s)
Annexin A7/physiology , Erythrocytes/physiology , Animals , Cell Death , Chlorides/physiology , Energy Metabolism , Mice , Mice, Knockout , Osmotic Pressure
11.
Nephrol Dial Transplant ; 25(9): 2938-44, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20368307

ABSTRACT

BACKGROUND: The mammalian target of rapamycin (mTOR) is known to stimulate a variety of transport mechanisms including the intestinal phosphate transporter NaPi-IIb. The present study was performed to elucidate whether mTOR similarly regulates the major renal tubular phosphate transporter NaPi-IIa. METHODS: To this end, NaPi-IIa was expressed in Xenopus oocytes with or without mTOR and phosphate transport estimated from phosphate-induced (1 mM) current (I(pi)). RESULTS: As a result, I(pi) was observed in NaPi-IIa-expressing but not in H(2)O-injected Xenopus oocytes. Co-expression of mTOR significantly enhanced I(pi) in NaPi-IIa-expressing Xenopus oocytes, an effect abrogated by treatment with rapamycin (50 nM for the last 24 h of incubation). In a second series of experiments, the effect of rapamycin was analysed in mice. The in vivo administration of rapamycin (3 microg/g body weight/day) for 3 days resulted in phosphaturia in mice despite a tendency of plasma phosphate concentration to decrease. CONCLUSIONS: mTOR contributes to the regulation of renal phosphate transport, and rapamycin thus influences phosphate balance.


Subject(s)
Hypophosphatemia, Familial/chemically induced , Immunosuppressive Agents/toxicity , Kidney/drug effects , Oocytes/drug effects , Phosphates/urine , Sirolimus/toxicity , TOR Serine-Threonine Kinases/metabolism , Animals , Biological Transport , Female , Hypophosphatemia, Familial/metabolism , Immunoenzyme Techniques , Kidney/metabolism , Male , Mice , Oocytes/cytology , Oocytes/metabolism , Rats , Sodium-Phosphate Cotransporter Proteins, Type IIa/metabolism , TOR Serine-Threonine Kinases/genetics , Xenopus laevis
12.
Am J Physiol Renal Physiol ; 298(5): F1113-7, 2010 May.
Article in English | MEDLINE | ID: mdl-20164156

ABSTRACT

Akt/PKB is known to regulate the facilitative glucose carrier GLUT4. Nothing is known, however, of the role of Akt/PKB in the regulation of renal epithelial transport. To explore whether Akt2/PKBß influences the Na(+)-coupled glucose cotransporter SGLT1, human SGLT1 was expressed in Xenopus laevis oocytes with or without Akt/PKB, and electrogenic glucose transport was determined by dual-electrode voltage clamp. The coexpression of Akt/PKB in SGLT1-expressing oocytes was followed by an increase in glucose-induced currents. To study the functional significance of Akt/PKB-sensitive renal glucose transport, further experiments were performed in gene-targeted mice lacking functional Akt2/PKBß (akt2(-/-)) and in their wild-type littermates (akt2(+/+)). Plasma glucose concentration was significantly higher in akt2(-/-) mice than in akt2(+/+) mice but was virtually identical to the plasma glucose concentration in fructose-treated akt2(+/+) mice. Urinary glucose excretion was significantly higher in akt2(-/-) mice compared with akt2(+/+) mice with or without fructose treatment. Moreover, the glucose-induced depolarization of proximal tubular cells was significantly smaller in isolated, perfused renal tubules from akt2(-/-) mice than in those from akt2(+/+) mice. In conclusion, Akt2/PKBß plays a role in the regulation of renal glucose transport.


Subject(s)
Glucose/metabolism , Kidney Tubules, Proximal/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sodium-Glucose Transporter 1/metabolism , Absorption , Animals , Female , Humans , Mice , Mice, Knockout , Models, Animal , Oocytes/cytology , Oocytes/metabolism , Patch-Clamp Techniques , Proto-Oncogene Proteins c-akt/deficiency , Proto-Oncogene Proteins c-akt/genetics , Xenopus laevis
13.
Cell Physiol Biochem ; 25(2-3): 203-10, 2010.
Article in English | MEDLINE | ID: mdl-20110681

ABSTRACT

Intestinal Na(+)-coupled glucose transporter SGLT1 determines the rate of glucose transport, which in turn influences glucose-induced insulin release and development of obesity. The present study explored effects of Gum Arabic (GA), a dietary polysaccharide from dried exudates of Acacia Senegal, on intestinal glucose transport and body weight in wild-type C57Bl/6 mice. Treatment with GA (100 g/l) in drinking water for four weeks did not affect intestinal SGLT1 transcript levels but decreased SGLT1 protein abundance in jejunal brush border membrane vesicles. Glucose-induced jejunal short-circuit currents revealed that GA treatment decreased electrogenic glucose transport. Drinking a 20% glucose solution for four weeks significantly increased body weight and fasting plasma glucose concentrations, effects significantly blunted by simultaneous treatment with GA. GA further significantly blunted the increase in body weight, fasting plasma glucose and fasting insulin concentrations during high fat diet. In conclusion, the present observations disclose a completely novel effect of gum arabic, i.e. its ability to decrease intestinal SGLT1 expression and activity and thus to counteract glucose-induced obesity.


Subject(s)
Gum Arabic/pharmacology , Jejunum/metabolism , Sodium-Glucose Transporter 1/metabolism , Sodium/metabolism , Animals , Down-Regulation , Glucose/metabolism , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Sodium-Glucose Transporter 1/genetics
14.
J Neuroimmunol ; 219(1-2): 81-9, 2010 Feb 26.
Article in English | MEDLINE | ID: mdl-20060598

ABSTRACT

Amyloid peptides are formed during inflammation and modify the function of immune cells. The present study explored the effect of amyloid beta-peptide (Abeta(1-42)) and islet amyloid polypeptide (IAPP) on bone marrow derived dendritic cells (DCs). DCs were treated with Abeta(1-42) or IAPP with subsequent assessment of ceramide formation, caspase 8 and 3 activity, DNA fragmentation and phosphatidylserine exposure. In addition, TNFalpha secretion was assessed in lypopolysaccharide (LPS)-stimulated Abeta(1-42)- or IAPP-treated DCs. Within 24h Abeta(1-42) and IAPP triggered ceramide formation, caspase 8 and caspase 3 activation, DNA fragmentation and annexin V binding in DCs obtained from wild type mice, whereas in DCs from sphingomyelinase deficient (asm(-/-)) mice and in wild type DCs treated with sphingomyelinase inhibitor amitriptyline all these effects were strongly impaired. Moreover, ceramide formation was also reduced in wild type DCs in which acid sphingomyelinase (Asm) was silenced with Asm-targeted siRNA. Finally, Abeta(1-42) and IAPP treatment was further followed by a decline of TNFalpha formation in wild type DCs. In conclusion, amyloid peptides induce DC apoptosis presumably through activation of acid sphingomyelinase resulting in production of ceramide.


Subject(s)
Amyloid beta-Peptides/pharmacology , Amyloid/pharmacology , Apoptosis/drug effects , Dendritic Cells/drug effects , Peptide Fragments/pharmacology , Sphingomyelin Phosphodiesterase/metabolism , Analysis of Variance , Animals , Annexin A5/metabolism , Bone Marrow , Caspase 3/metabolism , Caspase 8/metabolism , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Islet Amyloid Polypeptide , Lipopolysaccharides/pharmacology , Mice , Mice, Knockout , RNA, Small Interfering/pharmacology , Sphingomyelin Phosphodiesterase/deficiency , Tumor Necrosis Factor-alpha/metabolism
15.
Cell Physiol Biochem ; 24(5-6): 415-28, 2009.
Article in English | MEDLINE | ID: mdl-19910682

ABSTRACT

The course of malaria does not only depend on the virulence of the parasite Plasmodium but also on properties of host erythrocytes. Here, we show that infection of erythrocytes from human sickle cell trait (HbA/S) carriers with ring stages of P. falciparum led to significantly enhanced PGE(2) formation, Ca(2+) permeability, annexin-A7 degradation, phosphatidylserine (PS) exposure at the cell surface, and clearance by macrophages. P. berghei-infected erythrocytes from annexin-A7-deficient (annexin-A7(-/-)) mice were more rapidly cleared than infected wildtype cells. Accordingly, P. berghei-infected annexin-A7(-/-) mice developed less parasitemia than wildtype mice. The cyclooxygenase inhibitor aspirin decreased erythrocyte PS exposure in infected annexin-A7(-/-) mice and abolished the differences of parasitemia and survival between the genotypes. Conversely, the PGE(2)-agonist sulprostone decreased parasitemia and increased survival of wild type mice. In conclusion, PS exposure on erythrocytes results in accelerated clearance of Plasmodium ring stage-infected HbA/S or annexin-A7(-/-) erythrocytes and thus confers partial protection against malaria in vivo.


Subject(s)
Annexin A7/metabolism , Erythrocytes/metabolism , Plasmodium falciparum/physiology , Sickle Cell Trait/parasitology , Animals , Annexin A7/deficiency , Annexin A7/genetics , Aspirin/therapeutic use , Calcium/metabolism , Dinoprostone/analogs & derivatives , Dinoprostone/metabolism , Dinoprostone/therapeutic use , Erythrocytes/parasitology , Genotype , Hemoglobin A/metabolism , Hemoglobin, Sickle/metabolism , Humans , Mice , Mice, Knockout , Parasitemia/drug therapy , Phagocytosis , Phosphatidylserines/metabolism , Plasmodium berghei/growth & development , Plasmodium berghei/physiology , Plasmodium falciparum/growth & development , Sickle Cell Trait/metabolism
16.
Pflugers Arch ; 459(1): 191-202, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19672619

ABSTRACT

According to recent in vitro experiments, the peptide transporter PepT2 is stimulated by the serum- and glucocorticoid-inducible kinase SGK1. The present study explored the contribution of SGK1 to the regulation of electrogenic intestinal peptide transport. Intestinal PepT1 was expressed in Xenopus oocytes, and peptide transport was determined by dual electrode voltage clamping. Peptide transport in intestinal segments was determined utilizing Ussing chamber. Cytosolic pH (pH( i )) was determined by BCECF fluorescence and Na(+)/H(+) exchanger activity was estimated from Na(+)-dependent pH recovery (pH ( i )) following an ammonium pulse. In PepT1-expressing Xenopus oocytes, coexpression of SGK1 enhanced electrogenic peptide transport. Intestinal transport and pH( i ) of untreated mice were similar in SGK1 knockout mice (sgk1 ( -/- )) and their wild-type littermates (sgk1 ( +/+ )). Glucocorticoid treatment (4 days 10 microg/g body weight (bw)/day dexamethasone) increased peptide transport in sgk1 ( +/+ ) but not in sgk1 (-/-) mice. Irrespective of dexamethasone treatment, luminal peptide (5 mM glycyl-glycine) led to a similar early decrease of pH( i ) in sgk1 (-/-) and sgk1 (+/+) mice, but to a more profound and sustained decline of pH( i ) in sgk1 (-/-) than in sgk1 ( +/+ ) mice. In the presence and absence of glycyl-glycine, pH ( i ) was significantly enhanced by dexamethasone treatment in sgk1 ( +/+ ) mice, an effect significantly blunted in sgk1 ( -/- ) mice. During sustained exposure to glycyl-glycine, pH ( i ) was significantly larger in sgk1 (+/+) mice than in sgk1 (-/-) mice, irrespective of dexamethasone treatment. In conclusion, basal intestinal peptide transport does not require stimulation by SGK1. Glucocorticoid treatment stimulates both Na(+)/H(+) exchanger activity and peptide transport, effects partially dependent on SGK1. Moreover, chronic exposure to glycyl-glycine stimulates Na(+)/H(+) exchanger activity, an effect again involving SGK1.


Subject(s)
Dexamethasone/metabolism , Glucocorticoids/metabolism , Immediate-Early Proteins/metabolism , Intestinal Mucosa/metabolism , Protein Serine-Threonine Kinases/metabolism , Symporters/metabolism , Animals , Dipeptides/metabolism , Fluorescent Antibody Technique , Glycylglycine/metabolism , Hydrogen-Ion Concentration , Intestines/drug effects , Mice , Mice, Knockout , Patch-Clamp Techniques , Reverse Transcriptase Polymerase Chain Reaction , Sodium-Hydrogen Exchangers/metabolism , Xenopus
17.
Cell Physiol Biochem ; 24(3-4): 193-200, 2009.
Article in English | MEDLINE | ID: mdl-19710534

ABSTRACT

PPARgamma agonists, such as pioglitazone, are widely used in the treatment of diabetes and several further disorders. They enhance transcription of the serum and glucocorticoid inducible kinase SGK1, which could in turn enhance gastric acid secretion by stimulating KCNQ1 K+ channels. The present study explored whether pioglitazone upregulates SGK1 protein expression in gastric glands and thus modifies gastric acid secretion. Food containing the PPARgamma agonist pioglitazone (approximately 25 mg/kg bw/day) was administered to gene-targeted mice lacking SGK1 (sgk1-/-, n=11) and their wild-type littermates (sgk1+/+, n=11). Western blotting was employed to analyze SGK1 expression, BCECF-fluorescence to determine acid secretion in isolated gastric glands and immunohistochemistry to elucidate KCNQ1 and H+/K+-ATPase protein abundance in the parietal cell membrane. Pioglitazone significantly increased SGK1 expression. Cytosolic pH and cellular buffer capacity were not significantly different between sgk1+/+ and sgk1-/- mice and not significantly modified in either genotype by pioglitazone. Without pioglitazone treatment, Na+-independent pH-recovery following an ammonium pulse (DeltapH/min) reflecting H+/K+-ATPase activity was again similar in sgk1+/+ and sgk1-/- mice. Pioglitazone significantly increased DeltapH/min (approximately 3 fold) in sgk1+/+ but not in sgk1-/- mice. H+/K+-ATPase inhibitor omeprazole (100 microM) abolished DeltapH/min in both genotypes irrespective of pioglitazone treatment. Increase in local K+ concentrations to 35 mM (replacing Na+/NMDG) significantly increased DeltapH/min and abrogated the differences between genotypes. KCNQ1 and H+/K+-ATPase protein abundance in the parietal cell membrane was enhanced by pioglitazone treatment in sgk1+/+ but not in sgk1-/- mice. In conclusion, pioglitazone increases gastric acid secretion, an effect at least partially due to stimulation of SGK1 expression and SGK1-dependent upregulation of KCNQ1.


Subject(s)
Gastric Acid/metabolism , Gastric Mucosa/metabolism , Hypoglycemic Agents/pharmacology , Immediate-Early Proteins/deficiency , Protein Serine-Threonine Kinases/deficiency , Thiazolidinediones/pharmacology , Animals , Enzyme Inhibitors/pharmacology , Fluorescent Antibody Technique , Gastric Mucosa/drug effects , Gene Expression/drug effects , Hydrogen-Ion Concentration/drug effects , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Immunohistochemistry , KCNQ1 Potassium Channel/metabolism , Mice , Mice, Knockout , Omeprazole/pharmacology , PPAR gamma/agonists , Parietal Cells, Gastric/metabolism , Pioglitazone , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , Up-Regulation/drug effects
18.
Am J Physiol Regul Integr Comp Physiol ; 297(3): R571-5, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19494170

ABSTRACT

Adenomatous polyposis coli (APC) fosters degradation of beta-catenin, a multifunctional protein upregulating the serum- and glucocorticoid-inducible kinase (SGK1). SGK1 regulates a wide variety of renal transport processes. The present study explored the possibility that APC influences renal function. To this end, metabolic cage experiments were performed in mice carrying a loss-of-function mutation in the APC gene (apc(Min/+)), their wild-type littermates (apc(+/+)), and apc(Min/+) mice lacking functional SGK1 (apc(Min/+)/sgk1(-/-)). As a result, mean body weight, food intake, fluid intake, salt appetite, urinary flow, as well as plasma Na(+) and K(+) concentrations were similar in apc(Min/+) mice, apc(+/+) mice, and apc(Min/+)/sgk1(-/-) mice. Glomerular filtration rate and absolute renal Na(+) excretion were decreased, and fractional urinary K(+) excretion was enhanced in apc(Min/+) mice. The antinatriuresis, but not the hypofiltration and kaliuresis was partially reversed by additional lack of SGK1. Plasma corticosterone and aldosterone concentrations were significantly enhanced in apc(Min/+) mice. While the plasma corticosterone concentration was similar in apc(+/+) mice and apc(Min/+)/sgk1(-/-) mice, plasma aldosterone was even higher in apc(Min/+)/sgk1(-/-) mice than in apc(Min/+) mice. The hyperaldosteronism of apc(Min/+) mice was paralleled by significantly elevated plasma volume and blood pressure. The experiments reveal an influence of defective APC on adrenal hormone release and renal function, effects partially but not completely explained by APC dependence of SGK1 expression.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Blood Pressure , Blood Volume , Hyperaldosteronism/physiopathology , Hypertension/physiopathology , Kidney/physiopathology , Mutation , Adenomatous Polyposis Coli Protein/deficiency , Adenomatous Polyposis Coli Protein/genetics , Aldosterone/blood , Animals , Blood Pressure/genetics , Blood Volume/genetics , Body Weight , Corticosterone/blood , Drinking , Eating , Glomerular Filtration Rate , Homeostasis , Hyperaldosteronism/genetics , Hyperaldosteronism/metabolism , Hypertension/genetics , Hypertension/metabolism , Immediate-Early Proteins/deficiency , Immediate-Early Proteins/genetics , Kidney/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Natriuresis , Potassium/blood , Potassium/urine , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Sodium/blood , Sodium/urine , Sodium Chloride, Dietary/administration & dosage , Urodynamics , Wnt Proteins/metabolism
19.
Am J Physiol Regul Integr Comp Physiol ; 296(6): R1695-701, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19369585

ABSTRACT

The pore-forming K+-channel alpha-subunit KCNQ1 is expressed in a wide variety of tissues including heart, skeletal muscle, liver, and epithelia. Most recent evidence revealed an association of the KCNQ1 gene with the susceptibility to type 2 diabetes. KCNQ1 participates in the regulation of cell volume, which is, in turn, critically important for the regulation of metabolism by insulin. The present study explored the influence of KCNQ1 on insulin-induced cellular K+ uptake and glucose metabolism. Insulin (100 nM)-induced K+ uptake was determined in isolated perfused livers from KCNQ1-deficient mice (kcnq1(-/-)) and their wild-type littermates (kcnq1(+/+)). Moreover, plasma glucose and insulin levels, intraperitoneal glucose (3 g/kg) tolerance, insulin (0.15 U/kg)-induced hypoglycemia, and peripheral uptake of radiolabeled 3H-deoxy-glucose were determined in both genotypes. Insulin-stimulated hepatocellular K+ uptake was significantly more sustained in isolated perfused livers from kcnq1(-/-) mice than from kcnq1(+/+)mice. The decline of plasma glucose concentration following an intraperitoneal injection of insulin was again significantly more sustained in kcnq1(-/-) than in kcnq1(+/+) mice. Both fasted and nonfasted plasma glucose and insulin concentrations were significantly lower in kcnq1(-/-) than in kcnq1(+/+)mice. Following an intraperitoneal glucose injection, the peak plasma glucose concentration was significantly lower in kcnq1(-/-) than in kcnq1(+/+)mice. Uptake of 3H-deoxy-glucose into skeletal muscle, liver, kidney and lung tissue was significantly higher in kcnq1(-/-) than in kcnq1(+/+)mice. In conclusion, KCNQ1 counteracts the stimulation of cellular K+ uptake by insulin and thereby influences K+-dependent insulin signaling on glucose metabolism. The observations indicate that KCNQ1 is a novel molecule affecting insulin sensitivity of glucose metabolism.


Subject(s)
Glucose/metabolism , Insulin/metabolism , KCNQ1 Potassium Channel/deficiency , Liver/metabolism , Potassium/metabolism , Animals , Blood Glucose/metabolism , Deoxyglucose/metabolism , Female , Glucose/administration & dosage , Glucose Tolerance Test , Glucose Transporter Type 1/metabolism , Injections, Intraperitoneal , Insulin/administration & dosage , Insulin/blood , KCNQ1 Potassium Channel/genetics , Kidney/metabolism , Lung/metabolism , Male , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Signal Transduction , Time Factors
20.
Pflugers Arch ; 458(3): 503-12, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19184092

ABSTRACT

Klotho, a membrane protein mainly expressed in parathyroid glands, kidney, and choroid plexus, counteracts aging and increases the life span. Accordingly, life span is significantly shorter in Klotho-deficient mice (klotho(-/-)) than in their wild-type littermates (klotho(+/+)). The pleotropic effects of Klotho include inhibition of 1,25-dihydroxyvitamin D(3)(1,25(OH)(2)D(3)) formation. Vitamin D-deficient diet reverses the shortening of life span in klotho(-/-) mice. In a variety of cells, 1,25(OH)(2)D(3) stimulates Ca(2+) entry. In erythrocytes, increased Ca(2+) entry stimulates suicidal erythrocyte death, which is characterized by cell shrinkage and phosphatidylserine exposure at the erythrocyte surface. The present study explored the putative impact of Klotho on eryptosis. According to Fluo3 fluorescence, cytosolic Ca(2+) concentration was significantly larger in klotho(-/-) erythrocytes as compared to klotho(+/+) erythrocytes. According to annexin V-binding, phosphatidylserine exposure was significantly enhanced, and according to forward scatter, cell volume significantly decreased in klotho(-/-) erythrocytes as compared to klotho(+/+) erythrocytes. Energy depletion (13 h glucose depletion) and oxidative stress (35 min 1 mM tert-butyl-hydroxyl-peroxide [tert-BOOH]) increased phosphatidylserine exposure to values again significantly larger in klotho(-/-) erythrocytes as compared to klotho(+/+) erythrocytes. Reticulocyte number was significantly increased in klotho (-/-) mice, pointing to enhanced erythrocyte turnover. Vitamin D-deficient diet reversed the enhanced Ca(2+) entry and annexin V-binding of klotho(-/-) erythrocytes. The present observations reveal a novel function of Klotho, i.e., the at least partially vitamin D-dependent regulation of cytosolic Ca(2+) activity in and suicidal death of erythrocytes.


Subject(s)
Calcium/metabolism , Erythrocytes/cytology , Erythrocytes/physiology , Glucuronidase/metabolism , Animals , Apoptosis/physiology , Calcium Signaling , Cell Death/physiology , Cells, Cultured , Female , Klotho Proteins , Male , Mice , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...