Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Am J Physiol Renal Physiol ; 322(3): F360-F377, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35073212

ABSTRACT

The seminal studies conducted by Giebisch and coworkers in the 1960s paved the way for understanding the renal mechanisms involved in K+ homeostasis. It was demonstrated that differential handling of K+ in the distal segments of the nephron is crucial for proper K+ balance. Although aldosterone had been classically ascribed as the major ion transport regulator in the distal nephron, thereby contributing to K+ homeostasis, it became clear that aldosterone per se could not explain the ability of the kidney to modulate kaliuresis in both acute and chronic settings. The existence of alternative kaliuretic and antikaliuretic mechanisms was suggested by physiological studies in the 1980s but only gained form and shape with the advent of molecular biology. It is now established that the kidneys recruit several endocrine and paracrine mechanisms for adequate kaliuretic response. These mechanisms include the direct effects of peritubular K+, a gut-kidney regulatory axis sensing dietary K+ levels, the kidney secretion of kallikrein during postprandial periods, the upregulation of angiotensin II receptors in the distal nephron during chronic changes in K+ diet, and the local increase of prostaglandins by low-K+ diet. This review discusses recent advances in the understanding of endocrine and paracrine mechanisms underlying the modulation of K+ secretion and how these mechanisms impact kaliuresis and K+ balance. We also highlight important unknowns about the regulation of renal K+ excretion under physiological circumstances.


Subject(s)
Aldosterone , Potassium , Aldosterone/pharmacology , Homeostasis , Kidney , Nephrons , Potassium/pharmacology
2.
J Am Soc Nephrol ; 32(7): 1616-1629, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33846238

ABSTRACT

BACKGROUND: SGLT2 inhibitors reduce the risk of heart failure (HF) mortality and morbidity, regardless of the presence or absence of diabetes, but the mechanisms underlying this benefit remain unclear. Experiments with nondiabetic HF rats tested the hypothesis that the SGLT2 inhibitor empagliflozin (EMPA) inhibits proximal tubule (PT) NHE3 activity and improves renal salt and water handling. METHODS: Male Wistar rats were subjected to myocardial infarction or sham operation. After 4 weeks, rats that developed HF and sham rats were treated with EMPA or untreated for an additional 4 weeks. Immunoblotting and quantitative RT-PCR evaluated SGLT2 and NHE3 expression. Stationary in vivo microperfusion measured PT NHE3 activity. RESULTS: EMPA-treated HF rats displayed lower serum B-type natriuretic peptide levels and lower right ventricle and lung weight to tibia length than untreated HF rats. Upon saline challenge, the diuretic and natriuretic responses of EMPA-treated HF rats were similar to those of sham rats and were higher than those of untreated HF rats. Additionally, EMPA treatment prevented GFR decline and renal atrophy in HF rats. PT NHE3 activity was higher in HF rats than in sham rats, whereas treatment with EMPA markedly reduced NHE3 activity. Unexpectedly, SGLT2 protein and mRNA abundance were upregulated in the PT of HF rats. CONCLUSIONS: Prevention of HF progression by EMPA is associated with reduced PT NHE3 activity, restoration of euvolemia, and preservation of renal mass. Moreover, dysregulation of PT SGLT2 may be involved in the pathophysiology of nondiabetic HF.

3.
Kidney Blood Press Res ; 42(6): 1277-1289, 2017.
Article in English | MEDLINE | ID: mdl-29262407

ABSTRACT

BACKGROUND/AIMS: To assess the possible contribution of the ß-adrenergic overstimulation in early stages of renal injury, the present study evaluated, in rats, the effects of the ß-adrenoceptor agonist isoproterenol (ISO) on renal function and morphology, as well as the renal mRNA and protein expression of the NADPH oxidase isoform 4 (Nox 4) and subunit p22phox, endoplasmic reticulum (ER) stress, pro-inflammatory, pro-apoptotic and renin-angiotensin system (RAS) components. METHODS: Wistar rats received ISO (0.3 mg.kg-1.day-1 s.c.) or vehicle (control) for eight days. At the end of the treatment, food and water intake, urine output and body weight gain were evaluated and renal function studies were performed. Renal tissue was used for the morphological, quantitative PCR and immunohistochemical studies. RESULTS: ISO did not change metabolic parameters or urine output. However it induced a decrease in renal blood flow and an increase in the filtration fraction. These changes were accompanied by increased cortical mRNA and protein expression for the renal oxidative stress components including Nox 4 and p22phox; ER stress, pro-inflamatory, pro-apoptotic as well as RAS components. ISO also induced a significant increase in medullar renin protein expression. CONCLUSION: These findings support relevant information regarding the contribution of specific ß-adrenergic hyperactivity in early stage of renal injury, indicating the reactive oxygen species, ER stress and intrarenal RAS as important factors in this process.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Kidney/injuries , Animals , Endoplasmic Reticulum Stress , Isoproterenol/pharmacology , Kidney Function Tests , Rats , Rats, Wistar , Reactive Oxygen Species , Renin-Angiotensin System
4.
Am J Physiol Renal Physiol ; 313(2): F450-F460, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28490531

ABSTRACT

The acute effects of angiotensin-1-7 [ANG-(1-7)] on the reabsorptive bicarbonate flow (J[Formula: see text]) were evaluated using stationary microperfusion in vivo in the proximal tubules of spontaneously hypertensive rats (SHR) and their normotensive controls, Wistar-Kyoto (WKY) rats, using a microelectrode sensitive to H+ In WKY rats, the control J[Formula: see text] was 2.40 ± 0.10 nmol·cm-2·s-1 (n = 120); losartan (10-7 M) or A779 (10-6 M, a specific Mas antagonist), alone or in combination with losartan, decreased the J[Formula: see text] ANG-(1-7) had biphasic effects on J[Formula: see text]: at 10-9 M, it inhibited, and at 10-6, it stimulated the flow. S3226 [10-6 M, a specific Na+-H+ exchanger 3 (NHE3) antagonist] decreased J[Formula: see text] and changed the stimulatory effect of ANG-(1-7) to an inhibitory one but did not alter the inhibitory action of ANG-(1-7). In SHR, the control J[Formula: see text] was 2.04 ± 0.13 nmol·cm-2·s-1 (n = 56), and A779 and/or losartan reduced the flow. ANG-(1-7) at 10-9 M increased J[Formula: see text], and ANG-(1-7) at 10-6 M reduced it. The effects of A779, losartan, and S3226 on the J[Formula: see text] were similar to those found in WKY rats, which indicated that in SHR, the ANG-(1-7) action on the NHE3 was via Mas and ANG II type 1. The cytosolic calcium in the WKY or SHR rats was ~100 nM and was increased by ANG-(1-7) at 10-9 or 10-6 M. In hypertensive animals, a high plasma level of ANG-(1-7) inhibited NHE3 in the proximal tubule, which mitigated the hypertension caused by the high plasma level of ANG II.


Subject(s)
Angiotensin I/pharmacology , Bicarbonates/metabolism , Blood Pressure/drug effects , Calcium/metabolism , Hypertension/metabolism , Kidney Tubules, Proximal/drug effects , Peptide Fragments/pharmacology , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Angiotensin II/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Hypertension/physiopathology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/physiopathology , Male , Proto-Oncogene Mas , Proto-Oncogene Proteins/agonists , Proto-Oncogene Proteins/metabolism , Rats, Inbred SHR , Rats, Inbred WKY , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Renal Reabsorption/drug effects , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/metabolism
5.
Biochim Biophys Acta ; 1860(7): 1431-8, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27102282

ABSTRACT

BACKGROUND: The natriuretic effect of uroguanylin (UGN) involves reduction of proximal tubule (PT) sodium reabsorption. However, the target sodium transporters as well as the molecular mechanisms involved in these processes remain poorly understood. METHODS: To address the effects of UGN on PT (Na(+)+K(+))ATPase and the signal transduction pathways involved in this effect, we used LLC-PK1 cells. The effects of UGN were determined through ouabain-sensitive ATP hydrolysis and immunoblotting assays during different experimental conditions. RESULTS: We observed that UGN triggers cGMP/PKG and cAMP/PKA pathways in a sequential way. The activation of PKA leads to the inhibition of mTORC2 activity, PKB phosphorylation at S473, PKB activity and, consequently, a decrease in the mTORC1/S6K pathway. The final effects are decreased expression of the α1 subunit of (Na(+)+K(+))ATPase and inhibition of enzyme activity. CONCLUSIONS: These results suggest that the molecular mechanism of action of UGN on sodium reabsorption in PT cells is more complex than previously thought. We propose that PKG-dependent activation of PKA leads to the inhibition of the mTORC2/PKB/mTORC1/S6K pathway, an important signaling pathway involved in the maintenance of the PT sodium pump expression and activity. GENERAL SIGNIFICANCE: The current results expand our understanding of the signal transduction pathways involved in the overall effect of UGN on renal sodium excretion.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Kidney Tubules, Proximal/drug effects , Natriuretic Agents/pharmacology , Natriuretic Peptides/pharmacology , Second Messenger Systems/drug effects , Sodium-Potassium-Exchanging ATPase/metabolism , TOR Serine-Threonine Kinases/metabolism , Adenosine Triphosphate/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Dose-Response Relationship, Drug , Enzyme Activation , Hydrolysis , Kidney Tubules, Proximal/enzymology , LLC-PK1 Cells , Natriuresis/drug effects , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Renal Elimination/drug effects , Renal Reabsorption/drug effects , Sodium/metabolism , Swine , TOR Serine-Threonine Kinases/antagonists & inhibitors
6.
Am J Physiol Renal Physiol ; 310(2): F123-7, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26447224

ABSTRACT

Glucagon like peptide-1 (GLP-1) is an incretin hormone constantly secreted from the intestine at low basal levels in the fasted state; plasma concentrations rise rapidly after nutrient ingestion. Upon release, GLP-1 exerts insulinotropic effects via a G protein-coupled receptor, stimulation of adenylyl cyclase, and cAMP generation. Although primarily involved in glucose homeostasis, GLP-1 can induce diuresis and natriuresis when administered in pharmacological doses in humans and rodents. However, whether endogenous GLP-1 plays a role in regulating renal function remains an open question. This study aimed to test the hypothesis that blockade of GLP-1 receptor (GLP-1R) signaling at baseline influences renal salt and water handling. To this end, the GLP-1R antagonist exendin-9 (100 µg·kg(-1)·min(-1)) or vehicle was administered intravenously to overnight-fasted male Wistar rats for 30 min. This treatment reduced urinary cAMP excretion and renal cortical PKA activity, demonstrating blockade of renal GLP-1R signaling. Exendin-9-infused-rats exhibited reduced glomerular filtration rate, lithium clearance, urinary volume flow, and sodium excretion compared with vehicle-infused controls. Exendin-9 infusion also reduced renal cortical Na(+)/H(+) exchanger isotope 3 (NHE3) phosphorylation at serine 552 (NHE3pS552), a PKA consensus site that correlates with reduced transport activity. Collectively, these results provide novel evidence that GLP-1 is a physiologically relevant natriuretic factor that contributes to sodium balance, in part via tonic modulation of NHE3 activity in the proximal tubule.


Subject(s)
Glucagon-Like Peptide-1 Receptor/antagonists & inhibitors , Kidney/drug effects , Natriuresis/drug effects , Natriuretic Agents/pharmacology , Peptide Fragments/pharmacology , Animals , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Glomerular Filtration Rate/drug effects , Kidney/metabolism , Male , Phosphorylation/drug effects , Rats , Rats, Wistar , Signal Transduction/drug effects , Sodium-Hydrogen Exchangers/metabolism
7.
PLoS One ; 10(12): e0146042, 2015.
Article in English | MEDLINE | ID: mdl-26713849

ABSTRACT

The enterotoxigenic Escherichia coli strains lead to diarrhoea in humans due to heat-labile and heat-stable (STa) enterotoxins. STa increases Cl-release in intestinal cells, including the human colonic carcinoma T84 cell line, involving increased cGMP and membrane alkalization due to reduced Na+/H+ exchangers (NHEs) activity. Since NHEs modulate intracellular pH (pHi), and NHE1, NHE2, and NHE4 are expressed in T84 cells, we characterized the STa role as modulator of these exchangers. pHi was assayed by the NH4Cl pulse technique and measured by fluorescence microscopy in BCECF-preloaded cells. pHi recovery rate (dpHi/dt) was determined in the absence or presence of 0.25 µmol/L STa (30 minutes), 25 µmol/L HOE-694 (concentration inhibiting NHE1 and NHE2), 500 µmol/L sodium nitroprusside (SNP, spontaneous nitric oxide donor), 100 µmol/L dibutyryl cyclic GMP (db-cGMP), 100 nmol/L H89 (protein kinase A inhibitor), or 10 µmol/L forskolin (adenylyl cyclase activator). cGMP and cAMP were measured in cell extracts by radioimmunoassay, and buffering capacity (ßi) and H+ efflux (JH+) was determined. NHE4 protein abundance was determined by western blotting. STa and HOE-694 caused comparable reduction in dpHi/dt and JH+ (~63%), without altering basal pHi (range 7.144-7.172). STa did not alter ßi value in a range of 1.6 pHi units. The dpHi/dt and JH+ was almost abolished (~94% inhibition) by STa + HOE-694. STa effect was unaltered by db-cGMP or SNP. However, STa and forskolin increased cAMP level. STa-decreased dpHi/dt and JH+ was mimicked by forskolin, and STa + HOE-694 effect was abolished by H89. Thus, incubation of T84 cells with STa results in reduced NHE4 activity leading to a lower capacity of pHi recovery requiring cAMP, but not cGMP. STa effect results in a causal phenomenon (STa/increased cAMP/increased PKA activity/reduced NHE4 activity) ending with intracellular acidification that could have consequences in the gastrointestinal cells function promoting human diarrhoea.


Subject(s)
Cyclic AMP/metabolism , Enterotoxins/pharmacology , Epithelial Cells/drug effects , Escherichia coli , Hot Temperature , Intestines/cytology , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Adult , Cell Line, Tumor , Cyclic GMP/metabolism , Drug Stability , Enterotoxins/chemistry , Epithelial Cells/metabolism , Gene Expression Regulation/drug effects , Humans , Hydrogen-Ion Concentration , Kinetics , Male , Protons , Sodium-Hydrogen Exchangers/metabolism
8.
Am J Physiol Cell Physiol ; 309(8): C541-50, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26246427

ABSTRACT

Physiological concentrations of angiotensin II (ANG II) upregulate the activity of Na(+)/H(+) exchanger isoform 3 (NHE3) in the renal proximal tubule through activation of the ANG II type I (AT1) receptor/G protein-coupled signaling. This effect is key for maintenance of extracellular fluid volume homeostasis and blood pressure. Recent findings have shown that selective activation of the beta-arrestin-biased AT1 receptor signaling pathway induces diuresis and natriuresis independent of G protein-mediated signaling. This study tested the hypothesis that activation of this AT1 receptor/beta-arrestin signaling inhibits NHE3 activity in proximal tubule. To this end, we determined the effects of the compound TRV120023, which binds to the AT1R, blocks G-protein coupling, and stimulates beta-arrestin signaling on NHE3 function in vivo and in vitro. NHE3 activity was measured in both native proximal tubules, by stationary microperfusion, and in opossum proximal tubule (OKP) cells, by Na(+)-dependent intracellular pH recovery. We found that 10(-7) M TRV120023 remarkably inhibited proximal tubule NHE3 activity both in vivo and in vitro. Additionally, stimulation of NHE3 by ANG II was completely suppressed by TRV120023 both in vivo as well as in vitro. Inhibition of NHE3 activity by TRV120023 was associated with a decrease in NHE3 surface expression in OKP cells and with a redistribution from the body to the base of the microvilli in the rat proximal tubule. These findings indicate that biased signaling of the beta-arrestin pathway through the AT1 receptor inhibits NHE3 activity in the proximal tubule at least in part due to changes in NHE3 subcellular localization.


Subject(s)
Arrestins/metabolism , Kidney Tubules, Proximal/cytology , Receptor, Angiotensin, Type 1/physiology , Sodium-Hydrogen Exchangers/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Humans , Hydrogen-Ion Concentration , Kidney Tubules, Proximal/physiology , Male , Oligopeptides/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction , Sodium/metabolism , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/genetics , beta-Arrestins
9.
Am J Physiol Cell Physiol ; 307(6): C532-41, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25031022

ABSTRACT

Cumulative evidence suggests that guanylin peptides play an important role on electrolyte homeostasis. We have previously reported that uroguanylin (UGN) inhibits bicarbonate reabsorption in a renal distal tubule. In the present study, we tested the hypothesis that the bicarbonaturic effect of UGN is at least in part attributable to inhibition of H(+)-ATPase-mediated hydrogen secretion in the distal nephron. By in vivo stationary microperfusion experiments, we were able to show that UGN inhibits H(+)-ATPase activity by a PKG-dependent pathway because KT5823 (PKG inhibitor) abolished the UGN effect on distal bicarbonate reabsorption and H89 (PKA inhibitor) was unable to prevent it. The in vivo results were confirmed by the in vitro experiments, where we used fluorescence microscopy to measure intracellular pH (pHi) recovery after an acid pulse with NH4Cl. By this technique, we observed that UGN and 8 bromoguanosine-cGMP (8Br-cGMP) inhibited H(+)-ATPase-dependent pHi recovery and that the UGN inhibitory effect was abolished in the presence of the PKG inhibitor. In addition, by using RT-PCR technique, we verified that Madin-Darby canine kidney (MDCK)-C11 cells express guanylate cyclase-C. Besides, UGN stimulated an increase of both cGMP content and PKG activity but was unable to increase the production of cellular cAMP content and PKA activity. Furthermore, we found that UGN reduced cell surface abundance of H+-ATPase B1 subunit in MDCK-C11 and that this effect was abolished by the PKG inhibitor. Taken together, our data suggest that UGN inhibits H(+)-ATPase activity and surface expression in renal distal cells by a cGMP/PKG-dependent pathway.


Subject(s)
Cell Membrane/drug effects , Cyclic GMP-Dependent Protein Kinases/metabolism , Kidney Tubules, Distal/drug effects , Natriuretic Peptides/pharmacology , Proton-Translocating ATPases/metabolism , Animals , Bicarbonates/metabolism , Cell Membrane/enzymology , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Dogs , Hydrogen-Ion Concentration , Kidney Tubules, Distal/enzymology , Madin Darby Canine Kidney Cells , Male , Perfusion , Protein Kinase Inhibitors/pharmacology , Protein Transport , Rats , Rats, Wistar , Receptors, Guanylate Cyclase-Coupled/drug effects , Receptors, Guanylate Cyclase-Coupled/genetics , Receptors, Guanylate Cyclase-Coupled/metabolism , Signal Transduction/drug effects , Time Factors
10.
J Am Soc Nephrol ; 25(9): 2028-39, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24652792

ABSTRACT

Na(+)-glucose cotransporter 1 (SGLT1)-mediated glucose uptake leads to activation of Na(+)-H(+) exchanger 3 (NHE3) in the intestine by a process that is not dependent on glucose metabolism. This coactivation may be important for postprandial nutrient uptake. However, it remains to be determined whether SGLT-mediated glucose uptake regulates NHE3-mediated NaHCO3 reabsorption in the renal proximal tubule. Considering that this nephron segment also expresses SGLT2 and that the kidneys and intestine show significant variations in daily glucose availability, the goal of this study was to determine the effect of SGLT-mediated glucose uptake on NHE3 activity in the renal proximal tubule. Stationary in vivo microperfusion experiments showed that luminal perfusion with 5 mM glucose stimulates NHE3-mediated bicarbonate reabsorption. This stimulatory effect was mediated by glycolytic metabolism but not through ATP production. Conversely, luminal perfusion with 40 mM glucose inhibited NHE3 because of cell swelling. Notably, pharmacologic inhibition of SGLT activity by Phlorizin produced a marked inhibition of NHE3, even in the absence of glucose. Furthermore, immunofluorescence experiments showed that NHE3 colocalizes with SGLT2 but not SGLT1 in the rat renal proximal tubule. Collectively, these findings show that glucose exerts a bimodal effect on NHE3. The physiologic metabolism of glucose stimulates NHE3 transport activity, whereas, supraphysiologic glucose concentrations inhibit this exchanger. Additionally, Phlorizin-sensitive SGLT transporters and NHE3 interact functionally in the proximal tubule.


Subject(s)
Glucose/metabolism , Kidney Tubules, Proximal/metabolism , Sodium-Glucose Transporter 1/metabolism , Sodium-Glucose Transporter 2/metabolism , Sodium-Hydrogen Exchangers/metabolism , Adenosine Triphosphate/biosynthesis , Animals , Bicarbonates/metabolism , Bicarbonates/urine , Galactose/metabolism , Immunohistochemistry , Kidney Tubules, Proximal/drug effects , Male , Methylglucosides/metabolism , Models, Biological , Osmotic Pressure , Phlorhizin/pharmacology , Rats , Rats, Wistar , Sodium-Glucose Transporter 1/antagonists & inhibitors , Sodium-Glucose Transporter 2 Inhibitors , Sodium-Hydrogen Exchanger 3
11.
Kidney Blood Press Res ; 36(1): 320-34, 2012.
Article in English | MEDLINE | ID: mdl-23235337

ABSTRACT

BACKGROUND/AIMS: Fructose causes a sodium-sensitive hypertension and acutely reduces the urinary Na+ excretion, suggesting that it may regulate the activity of renal tubular sodium transporters. NHE3 is highly expressed in proximal tubule (PT), along with proteins that mediate fructose transport and metabolism. The present work was outlined to investigate whether fructose modulates proximal NHE3 activity and to elucidate the molecular mechanisms underlying this modulation. METHODS/RESULTS: Using in vivo stationary microperfusion, we observed that fructose stimulates NHE3 mediated JHCO3- reabsorption. The MAPK pathway is not involved in this activation, as demonstrated by using of MEK/MAPK inhibitors, whereas experiments using a PKA inhibitor suggest that PKA inhibition plays a role in this response. These results were confirmed in vitro by measuring the cell pH recovery rate after NH4Cl pulse in LLC-PK1, a pig PT cell line, which showed reduced cAMP levels and NHE3 phosphorylation at serine-552 (PKA consensus site) after fructose treatment. CONCLUSIONS: NHE3 activity is stimulated by fructose, which increases proximal tubule Na+ reabsorption. The molecular mechanisms involved in this process are mediated, at least in part, by downregulation of the PKA signaling pathway. Future studies are needed to address whether fructose-stimulated NHE3 activity may contribute to renal injury and hypertension.


Subject(s)
Fructose/pharmacology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Sodium-Hydrogen Exchangers/metabolism , Animals , Cell Line , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Fructokinases/metabolism , Glucose Transporter Type 2/metabolism , Glucose Transporter Type 5/metabolism , Kidney Tubules, Proximal/cytology , LLC-PK1 Cells , Male , Models, Animal , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology , Sodium-Hydrogen Exchanger 3 , Swine
12.
Am J Physiol Renal Physiol ; 303(10): F1399-408, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22952280

ABSTRACT

We previously demonstrated that uroguanylin (UGN) significantly inhibits Na(+)/H(+) exchanger (NHE)3-mediated bicarbonate reabsorption. In the present study, we aimed to elucidate the molecular mechanisms underlying the action of UGN on NHE3 in rat renal proximal tubules and in a proximal tubule cell line (LLC-PK(1)). The in vivo studies were performed by the stationary microperfusion technique, in which we measured H(+) secretion in rat renal proximal segments, through a H(+)-sensitive microelectrode. UGN (1 µM) significantly inhibited the net of proximal bicarbonate reabsorption. The inhibitory effect of UGN was completely abolished by either the protein kinase G (PKG) inhibitor KT5823 or by the protein kinase A (PKA) inhibitor H-89. The effects of UGN in vitro were found to be similar to those obtained by microperfusion. Indeed, we observed that incubation of LLC-PK(1) cells with UGN induced an increase in the intracellular levels of cAMP and cGMP, as well as activation of both PKA and PKG. Furthermore, we found that UGN can increase the levels of NHE3 phosphorylation at the PKA consensus sites 552 and 605 in LLC-PK(1) cells. Finally, treatment of LLC-PK(1) cells with UGN reduced the amount of NHE3 at the cell surface. Overall, our data suggest that the inhibitory effect of UGN on NHE3 transport activity in proximal tubule is mediated by activation of both cGMP/PKG and cAMP/PKA signaling pathways which in turn leads to NHE3 phosphorylation and reduced NHE3 surface expression. Moreover, this study sheds light on mechanisms by which guanylin peptides are intricately involved in the maintenance of salt and water homeostasis.


Subject(s)
Bicarbonates/metabolism , Kidney Tubules, Proximal/drug effects , Natriuretic Peptides/pharmacology , Sodium-Hydrogen Exchangers/metabolism , Animals , Carbazoles/pharmacology , Cell Line , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Isoquinolines/pharmacology , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Male , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Wistar , Sodium-Hydrogen Exchanger 3 , Sulfonamides/pharmacology
13.
Am J Physiol Regul Integr Comp Physiol ; 302(1): R166-74, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22031782

ABSTRACT

Heart failure (HF) is associated with a reduced effective circulating volume that drives sodium and water retention and extracellular volume expansion. We therefore hypothesized that Na(+)/H(+) exchanger isoform 3 (NHE3), the major apical transcellular pathway for sodium reabsorption in the proximal tubule, is upregulated in an experimental model of HF. HF was induced in male rats by left ventricle radiofrequency ablation. Sham-operated rats (sham) were used as controls. At 6 wk after surgery, HF rats exhibited cardiac dysfunction with a dramatic increase in left ventricular end-diastolic pressure. By means of stationary in vivo microperfusion and pH-dependent sodium uptake, we demonstrated that NHE3 transport activity was significantly higher in the proximal tubule of HF compared with sham rats. Increased NHE3 activity was paralleled by increased renal cortical NHE3 expression at both protein and mRNA levels. In addition, the baseline PKA-dependent NHE3 phosphorylation at serine 552 was reduced in renal cortical membranes of rats with HF. Collectively, these results suggest that NHE3 is upregulated in the proximal tubule of HF rats by transcriptional, translational, and posttranslational mechanisms. Enhanced NHE3-mediated sodium reabsorption in the proximal tubule may contribute to extracellular volume expansion and edema, the hallmark feature of HF. Moreover, our study emphasizes the importance of undertaking a cardiorenal approach to contain progression of cardiac disease.


Subject(s)
Heart Failure/metabolism , Kidney Tubules, Proximal/metabolism , Sodium-Hydrogen Exchangers/metabolism , Animals , Biological Transport , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Heart Failure/physiopathology , Kidney Tubules, Proximal/physiopathology , Male , Models, Animal , Phosphorylation , RNA, Messenger/metabolism , Rats , Rats, Wistar , Sodium-Hydrogen Exchanger 3
14.
Am J Physiol Renal Physiol ; 301(2): F355-63, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21593184

ABSTRACT

Glucagon-like peptide-1 (GLP-1) is a gut incretin hormone considered a promising therapeutic agent for type 2 diabetes because it stimulates beta cell proliferation and insulin secretion in a glucose-dependent manner. Cumulative evidence supports a role for GLP-1 in modulating renal function; however, the mechanisms by which GLP-1 induces diuresis and natriuresis have not been completely established. This study aimed to define the cellular and molecular mechanisms mediating the renal effects of GLP-1. GLP-1 (1 µg·kg(-1)·min(-1)) was intravenously administered in rats for the period of 60 min. GLP-1-infused rats displayed increased urine flow, fractional excretion of sodium, potassium, and bicarbonate compared with those rats that received vehicle (1% BSA/saline). GLP-1-induced diuresis and natriuresis were also accompanied by increases in renal plasma flow and glomerular filtration rate. Real-time RT-PCR in microdissected rat nephron segments revealed that GLP-1 receptor-mRNA expression was restricted to glomerulus and proximal convoluted tubule. In rat renal proximal tubule, GLP-1 significantly reduced Na(+)/H(+) exchanger isoform 3 (NHE3)-mediated bicarbonate reabsorption via a protein kinase A (PKA)-dependent mechanism. Reduced proximal tubular bicarbonate flux rate was associated with a significant increase of NHE3 phosphorylation at the PKA consensus sites in microvillus membrane vesicles. Taken together, these data suggest that GLP-1 has diuretic and natriuretic effects that are mediated by changes in renal hemodynamics and by downregulation of NHE3 activity in the renal proximal tubule. Moreover, our findings support the view that GLP-1-based agents may have a potential therapeutic use not only as antidiabetic drugs but also in hypertension and other disorders of sodium retention.


Subject(s)
Glucagon-Like Peptide 1/metabolism , Kidney/metabolism , Natriuretic Agents/administration & dosage , Animals , Cyclic AMP/urine , Cyclic AMP-Dependent Protein Kinases/metabolism , Exenatide , Glucagon-Like Peptide 1/administration & dosage , Glucagon-Like Peptide-1 Receptor , Hypoglycemic Agents/pharmacology , Kidney/drug effects , Natriuretic Agents/metabolism , Pentanoic Acids/pharmacology , Peptides/drug effects , Phosphorylation/drug effects , Rats , Rats, Wistar , Receptors, Glucagon/metabolism , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/metabolism , Thiazolidines/pharmacology , Venoms
15.
São Paulo; Atheneu; 2011. 408 p. ilus.
Monography in Portuguese | LILACS, HSPM-Producao, Sec. Munic. Saúde SP, Sec. Munic. Saúde SP | ID: lil-655143

ABSTRACT

O livro tem sua inspiração no Curso de Bases Fisiológicas de Prática Médica da Faculdade de Medicina da Universidade de São Paulo – USP, capítulo Nefrologia. Seu texto passou por sucessivas modificações didáticas e de conteúdo, ao longo dos 18 anos de existência do curso. Amadureceu, encorpou, ganhando aspecto interdisciplinar ao interagir a Fisiologia – centrada nos mecanismos de funcionamento de órgãos e sistemas, com a prática clínica dirigida à interpretação de dados clínico laboratoriais e a aplicação do raciocínio diagnóstico. É livro, pois, que bem reflete a excelência do ensino da Nefrologia integrado pela Fisiologia, Fisiopatologia Renal, Investigações clínicas e Exames Complementares. Sua equipe autorial é formada por 1 Editor, 2 Coeditores e 14 Colaboradores. Apresenta 15 capítulos e 3 Apêndices. É destinado aos estudantes de graduação de Medicina, Médicos Residentes e profissionais interessados em reciclar e atualizar seus conhecimentos em Nefrologia.


Subject(s)
Humans , Calcium/deficiency , Dehydration , Edema/physiopathology , Glomerular Filtration Rate , Hypertension/physiopathology , Potassium Deficiency , Renal Insufficiency
16.
In. Zatz, Roberto; Seguro, Antonio Carlos; Malnic, Gerhard. Bases fisiológicas da nefrologia. São Paulo, Atheneu, 2011. p.45-84, ilus.
Monography in Portuguese | LILACS, HSPM-Producao, Sec. Munic. Saúde SP, Sec. Munic. Saúde SP | ID: lil-655144
17.
São Paulo; Atheneu; 2011. 408 p. ilus.
Monography in Portuguese | LILACS, Coleciona SUS, HSPM-Producao, Sec. Munic. Saúde SP, Sec. Munic. Saúde SP | ID: biblio-938055

ABSTRACT

O livro tem sua inspiração no Curso de Bases Fisiológicas de Prática Médica da Faculdade de Medicina da Universidade de São Paulo – USP, capítulo Nefrologia. Seu texto passou por sucessivas modificações didáticas e de conteúdo, ao longo dos 18 anos de existência do curso. Amadureceu, encorpou, ganhando aspecto interdisciplinar ao interagir a Fisiologia – centrada nos mecanismos de funcionamento de órgãos e sistemas, com a prática clínica dirigida à interpretação de dados clínico laboratoriais e a aplicação do raciocínio diagnóstico. É livro, pois, que bem reflete a excelência do ensino da Nefrologia integrado pela Fisiologia, Fisiopatologia Renal, Investigações clínicas e Exames Complementares. Sua equipe autorial é formada por 1 Editor, 2 Coeditores e 14 Colaboradores. Apresenta 15 capítulos e 3 Apêndices. É destinado aos estudantes de graduação de Medicina, Médicos Residentes e profissionais interessados em reciclar e atualizar seus conhecimentos em Nefrologia


Subject(s)
Humans , Calcium/deficiency , Dehydration , Edema/physiopathology , Glomerular Filtration Rate , Hypertension/physiopathology , Potassium Deficiency , Renal Insufficiency
18.
São Paulo; Atheneu; 2011. 408 p. ilus.
Monography in Portuguese | Sec. Munic. Saúde SP, HSPM-Producao, Sec. Munic. Saúde SP, Sec. Munic. Saúde SP | ID: sms-4804

ABSTRACT

O livro tem sua inspiração no Curso de Bases Fisiológicas de Prática Médica da Faculdade de Medicina da Universidade de São Paulo – USP, capítulo Nefrologia. Seu texto passou por sucessivas modificações didáticas e de conteúdo, ao longo dos 18 anos de existência do curso. Amadureceu, encorpou, ganhando aspecto interdisciplinar ao interagir a Fisiologia – centrada nos mecanismos de funcionamento de órgãos e sistemas, com a prática clínica dirigida à interpretação de dados clínico laboratoriais e a aplicação do raciocínio diagnóstico. É livro, pois, que bem reflete a excelência do ensino da Nefrologia integrado pela Fisiologia, Fisiopatologia Renal, Investigações clínicas e Exames Complementares. Sua equipe autorial é formada por 1 Editor, 2 Coeditores e 14 Colaboradores. Apresenta 15 capítulos e 3 Apêndices. É destinado aos estudantes de graduação de Medicina, Médicos Residentes e profissionais interessados em reciclar e atualizar seus conhecimentos em Nefrologia


Subject(s)
Humans , Glomerular Filtration Rate , Dehydration , Edema/physiopathology , Hypertension/physiopathology , Potassium Deficiency , Calcium/deficiency , Renal Insufficiency
19.
In. Zatz, Roberto; Seguro, Antonio Carlos; Malnic, Gerhard. Bases fisiológicas da nefrologia. São Paulo, Atheneu, 2011. p.45-84, ilus.
Monography in Portuguese | Sec. Munic. Saúde SP, HSPM-Producao, Sec. Munic. Saúde SP, Sec. Munic. Saúde SP | ID: sms-4805
20.
J Nephrol ; 23 Suppl 16: S19-27, 2010.
Article in English | MEDLINE | ID: mdl-21170879

ABSTRACT

The functional versatility of the distal nephron is mainly due to the large cytological heterogeneity of the segment. Part of Na+ uptake by distal tubules is dependent on Na+/H+ exchanger 2 (NHE2), implicating a role of distal convoluted cells also in acid-base homeostasis. In addition, intercalated (IC) cells expressed in distal convoluted tubules, connecting tubules and collecting ducts are involved in the final regulation of acid-base excretion. IC cells regulate acid-base handling by 2 main transport proteins, a V-type H+-ATPase and a Cl/HCO3- exchanger, localized at different membrane domains. Type A IC cells are characterized by a luminal H+-ATPase in series with a basolateral Cl/HCO3- exchanger, the anion exchanger AE1. Type B IC cells mediate HCO3- secretion through the apical Cl-/HCO3- exchanger pendrin in series with a H+-ATPase at the basolateral membrane. Alternatively, H+/K+-ATPases have also been found in several distal tubule cells, particularly in type A and B IC cells. All of these mechanisms are finely regulated, and mutations of 1 or more proteins ultimately lead to expressive disorders of acid-base balance.


Subject(s)
Acid-Base Equilibrium/physiology , Kidney Tubules, Distal/metabolism , Nephrons/metabolism , Animals , Chloride-Bicarbonate Antiporters/physiology , H(+)-K(+)-Exchanging ATPase/physiology , Humans , Ion Transport , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/physiology , Vacuolar Proton-Translocating ATPases/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...