Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
J Am Heart Assoc ; 12(10): e026950, 2023 05 16.
Article in English | MEDLINE | ID: mdl-37183873

ABSTRACT

Background Cardiac metabolic abnormalities are present in heart failure. Few studies have followed metabolic changes accompanying diastolic and systolic heart failure in the same model. We examined metabolic changes during the development of diastolic and severe systolic dysfunction in spontaneously hypertensive rats (SHR). Methods and Results We serially measured myocardial glucose uptake rates with dynamic 2-[18F] fluoro-2-deoxy-d-glucose positron emission tomography in vivo in 9-, 12-, and 18-month-old SHR and Wistar Kyoto rats. Cardiac magnetic resonance imaging determined systolic function (ejection fraction) and diastolic function (isovolumetric relaxation time) and left ventricular mass in the same rats. Cardiac metabolomics was performed at 12 and 18 months in separate rats. At 12 months, SHR hearts, compared with Wistar Kyoto hearts, demonstrated increased isovolumetric relaxation time and slightly reduced ejection fraction indicating diastolic and mild systolic dysfunction, respectively, and higher (versus 9-month-old SHR decreasing) 2-[18F] fluoro-2-deoxy-d-glucose uptake rates (Ki). At 18 months, only few SHR hearts maintained similar abnormalities as 12-month-old SHR, while most exhibited severe systolic dysfunction, worsening diastolic function, and markedly reduced 2-[18F] fluoro-2-deoxy-d-glucose uptake rates. Left ventricular mass normalized to body weight was elevated in SHR, more pronounced with severe systolic dysfunction. Cardiac metabolite changes differed between SHR hearts at 12 and 18 months, indicating progressive defects in fatty acid, glucose, branched chain amino acid, and ketone body metabolism. Conclusions Diastolic and severe systolic dysfunction in SHR are associated with decreasing cardiac glucose uptake, and progressive abnormalities in metabolite profiles. Whether and which metabolic changes trigger progressive heart failure needs to be established.


Subject(s)
Heart Failure , Hypertension , Rats , Animals , Rats, Inbred SHR , Tomography, X-Ray Computed , Rats, Inbred WKY , Glucose , Deoxyglucose , Blood Pressure
2.
Circ Res ; 132(9): 1127-1140, 2023 04 28.
Article in English | MEDLINE | ID: mdl-36919600

ABSTRACT

BACKGROUND: Extracellular renal interstitial guanosine cyclic 3',5'-monophosphate (cGMP) inhibits renal proximal tubule (RPT) sodium (Na+) reabsorption via Src (Src family kinase) activation. Through which target extracellular cGMP acts to induce natriuresis is unknown. We hypothesized that cGMP binds to the extracellular α1-subunit of NKA (sodium-potassium ATPase) on RPT basolateral membranes to inhibit Na+ transport similar to ouabain-a cardiotonic steroid. METHODS: Urine Na+ excretion was measured in uninephrectomized 12-week-old female Sprague-Dawley rats that received renal interstitial infusions of vehicle (5% dextrose in water), cGMP (18, 36, and 72 µg/kg per minute; 30 minutes each), or cGMP+rostafuroxin (12 ng/kg per minute) or were subjected to pressure-natriuresis±rostafuroxin infusion. Rostafuroxin is a digitoxigenin derivative that displaces ouabain from NKA. RESULTS: Renal interstitial cGMP and raised renal perfusion pressure induced natriuresis and increased phosphorylated SrcTyr416 and Erk 1/2 (extracellular signal-regulated protein kinase 1/2)Thr202/Tyr204; these responses were abolished with rostafuroxin coinfusion. To assess cGMP binding to NKA, we performed competitive binding studies with isolated rat RPTs using bodipy-ouabain (2 µM)+cGMP (10 µM) or rostafuroxin (10 µM) and 8-biotin-11-cGMP (2 µM)+ouabain (10 µM) or rostafuroxin (10 µM). cGMP or rostafuroxin reduced bodipy-ouabain fluorescence intensity, and ouabain or rostafuroxin reduced 8-biotin-11-cGMP staining. We cross-linked isolated rat RPTs with 4-N3-PET-8-biotin-11-cGMP (2 µM); 8-N3-6-biotin-10-cAMP served as negative control. Precipitation with streptavidin beads followed by immunoblot analysis showed that RPTs after cross-linking with 4-N3-PET-8-biotin-11-cGMP exhibited a significantly stronger signal for NKA than non-cross-linked samples and cross-linked or non-cross-linked 8-N3-6-biotin-10-cAMP RPTs. Ouabain (10 µM) reduced NKA in cross-linked 4-N3-PET-8-biotin-11-cGMP RPTs confirming fluorescence staining. 4-N3-PET-8-biotin-11-cGMP cross-linked samples were separated by SDS gel electrophoresis and slices corresponding to NKA molecular weight excised and processed for mass spectrometry. NKA was the second most abundant protein with 50 unique NKA peptides covering 47% of amino acids in NKA. Molecular modeling demonstrated a potential cGMP docking site in the ouabain-binding pocket of NKA. CONCLUSIONS: cGMP can bind to NKA and thereby mediate natriuresis.


Subject(s)
Cyclic GMP , Natriuresis , Sodium-Potassium-Exchanging ATPase , Animals , Female , Rats , Adenosine Triphosphatases/metabolism , Biotin/metabolism , Cyclic GMP/chemistry , Cyclic GMP/metabolism , Natriuresis/physiology , Ouabain/pharmacology , Potassium/metabolism , Rats, Sprague-Dawley , Sodium/metabolism , Sodium-Potassium-Exchanging ATPase/chemistry , Sodium-Potassium-Exchanging ATPase/metabolism
3.
Circ Res ; 130(1): 96-111, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34794320

ABSTRACT

BACKGROUND: How signals from activated angiotensin type-2 receptors (AT2R) mediate inhibition of sodium ion (Na+) reabsorption in renal proximal tubule cells is currently unknown. Protein phosphatases including PP2A (protein phosphatase 2A) have been implicated in AT2R signaling in tissues other than kidney. We investigated whether inhibition of protein phosphatase PP2A reduced AT2R-mediated natriuresis and evaluated changes in PP2A activity and localization after renal AT2R activation in normal 4- and 10-week-old control Wistar-Kyoto rats and 4-week-old prehypertensive and 10-week-old hypertensive spontaneously hypertensive rats. METHODS AND RESULTS: In Wistar-Kyoto rats, direct renal interstitial administration of selective AT2R nonpeptide agonist Compound-21 (C-21) increased renal interstitial cyclic GMP (cGMP) levels, urine Na+ excretion, and simultaneously increased PP2A activity ≈2-fold in homogenates of renal cortical tubules. The cyclic GMP and natriuretic responses were abolished by concurrent renal interstitial administration of protein phosphatase inhibitor calyculin A. In renal proximal tubule cells in response to C-21, PP2A subunits A, B55α and C, but not B56γ, were recruited to apical plasma membranes together with AT2Rs. Calyculin A treatment abolished C-21-induced translocation of both AT2R and PP2A regulatory subunit B55α to apical plasma membranes. Immunoprecipitation of AT2R solubilized from renal cortical homogenates demonstrated physical association of AT2R with PP2A A, B55α, and C but not B56γ subunits. In contrast, in spontaneously hypertensive rats, administration of C-21 did not alter urine Na+ excretion or PP2A activity and failed to translocate AT2Rs and PP2A subunits to apical plasma membranes. CONCLUSIONS: In renal proximal tubule cells of Wistar-Kyoto rats, PP2A is activated and PP2A subunits AB55αC are recruited to C-21-activated AT2Rs during induction of natriuresis. This response is defective in prehypertensive and hypertensive spontaneously hypertensive rats, presenting a potential novel therapeutic target for treating renal Na+ retention and hypertension.


Subject(s)
Kidney/metabolism , Natriuresis , Protein Phosphatase 2/metabolism , Receptor, Angiotensin, Type 2/metabolism , Animals , Cells, Cultured , Cyclic GMP/metabolism , Female , Rats , Rats, Wistar , Sodium/metabolism
4.
Clin Sci (Lond) ; 134(7): 871-884, 2020 04 17.
Article in English | MEDLINE | ID: mdl-32202299

ABSTRACT

Recently, we designed a group of peptides by sequential substitution of the naturally occurring α-amino acid throughout the Ang III peptide sequence with the corresponding ß-amino acid. ß-Amino acid substitution at the proline residue of Ang III (ß-Pro7-Ang III) resulted in a highly selective AT2R ligand, demonstrating remarkable selectivity for the AT2R in both binding and functional studies. To provide additional functional evidence for the suitability of ß-Pro7 Ang III as a novel AT2R agonist, we tested effects of acute systemic administration of ß-Pro7-Ang III on renal hemodynamic and excretory function in anesthetized normotensive male and female rats. We also compared the natriuretic effects of acute intrarenal administration of native Ang III and ß-Pro7-Ang III in the presence of systemic AT1R blockade in anesthetized female rats to allow for the differentiation of systemic versus direct intrarenal natriuretic actions of ß-Pro7-Ang III. In both male and female rats, acute systemic administration of ß-Pro7-Ang III elicited renal vasodilatation and natriuresis. Notably, greater renal vasodilatory effects were observed in female versus male rats at the highest dose of ß-Pro7-Ang III administered. Moreover, intra-renal administration of ß-Pro7-Ang III produced significant natriuretic effects in female rats and, like Ang III, evoked AT2R translocation to the apical plasma membrane in renal proximal tubular cells. Taken together, our findings support the use of ß-Pro7-Ang III as a novel AT2R agonist and experimental tool for exploring AT2R function and its potential as a therapeutic target. Furthermore, our findings provide further evidence of a sex-specific influence of AT2R stimulation on renal function.


Subject(s)
Angiotensin III/analogs & derivatives , Kidney/blood supply , Kidney/drug effects , Natriuresis/drug effects , Receptor, Angiotensin, Type 2/agonists , Renal Circulation/drug effects , Renin-Angiotensin System/drug effects , Vasodilation/drug effects , Angiotensin III/pharmacology , Animals , Female , Kidney/metabolism , Male , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 2/metabolism , Sex Factors , Signal Transduction
5.
Circ Res ; 126(5): 644-659, 2020 02 28.
Article in English | MEDLINE | ID: mdl-31997705

ABSTRACT

RATIONALE: Previous studies identified a defect in Ang III (angiotensin III [des-aspartyl1-angiotensin II])-elicited AT2R (Ang type-2 receptor)-mediated natriuresis in renal proximal tubule cells of spontaneously hypertensive rats (SHR). OBJECTIVE: This study aimed to delineate in prehypertensive SHR kidneys the receptor or postreceptor defect causing impaired AT2R signaling and renal sodium (Na+) retention by utilizing the selective AT2R agonist compound-21 (C-21). METHODS AND RESULTS: Female 4-week-old Wistar Kyoto and SHR rats were studied after 24-hour systemic AT1R (Ang II type-1 receptor) blockade. Left kidneys received 30-minute renal interstitial infusions of vehicle followed by C-21 (20, 40, and 60 ng/[kg·min], each dose 30 minutes). Right kidneys received vehicle infusions. In Wistar Kyoto, C-21 dose-dependently increased urine Na+ excretion from 0.023±0.01 to 0.064±0.02, 0.087±0.01, and 0.089±0.01 µmol/min (P=0.008, P<0.0001, and P<0.0001, respectively) and renal interstitial fluid levels of AT2R downstream signaling molecule cGMP (cyclic guanosine 3',5' monophosphate) from 0.91±0.3 to 3.1±1.0, 5.9±1.2 and 5.3±0.5 fmol/mL (P=nonsignificant, P<0.0001, and P<0.0001, respectively). In contrast, C-21 did not increase urine Na+ excretion or renal interstitial cGMP in SHR. Mean arterial pressure was slightly higher in SHR but within the normotensive range and unaffected by C-21. In Wistar Kyoto, but not SHR, C-21 induced AT2R translocation to apical plasma membranes of renal proximal tubule cells, internalization/inactivation of NHE-3 (sodium-hydrogen exchanger-3) and Na+/K+ATPase (sodium-potassium-atpase) and phosphorylation of AT2R-cGMP downstream signaling molecules Src (Src family kinase), ERK (extracellular signal-related kinase), and VASP (vasodilator-stimulated phosphoprotein). To test whether cGMP could bypass the natriuretic defect in SHR, we infused 8-bromo-cGMP. This restored natriuresis, Na+ transporter internalization/inactivation, and Src and VASP phosphorylation, but not apical plasma membrane AT2R recruitment. In contrast, 8-bromo-cAMP administration had no effect on natriuresis or AT2R recruitment in SHR. CONCLUSIONS: The results demonstrate a primary renal proximal tubule cell AT2R natriuretic defect in SHR that may contribute to the development of hypertension. Since the defect is abrogated by exogenous intrarenal cGMP, the renal cGMP pathway may represent a viable target for the treatment of hypertension. Visual Overview: An online visual overview is available for this article.


Subject(s)
Hypertension/metabolism , Kidney Tubules, Proximal/metabolism , Natriuresis , Receptor, Angiotensin, Type 2/metabolism , Angiotensin II Type 2 Receptor Blockers/pharmacology , Animals , Cell Adhesion Molecules/metabolism , Cyclic GMP/metabolism , Extracellular Fluid/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Hypertension/genetics , Kidney Tubules, Proximal/drug effects , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Transport , Rats , Rats, Inbred SHR , Rats, Wistar , Sodium/metabolism , Sodium-Hydrogen Exchangers/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , src-Family Kinases/metabolism
6.
J Endocr Soc ; 3(11): 2088-2106, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31663064

ABSTRACT

Intrarenal ghrelin infusion activates ghrelin receptors in the kidney collecting duct (CD) to increase α epithelial sodium (Na+) channel (αENaC)-dependent Na+ reabsorption in vivo, but the underlying mechanisms are unknown. Seventy-two hours following uninephrectomy, 12-week-old female Sprague-Dawley rats received the following renal interstitial (RI) infusions for 1 hour after a 1-hour control: vehicle (n = 10), ghrelin (3 µg/minute; n = 8), ghrelin + phosphatidylinositol 3-kinase (PI3K) inhibitor LY-294002 (0.1 µg/kg/minute; n = 7), ghrelin + protein kinase A (PKA) inhibitor adenosine 3'5'-cyclic monophosphorothioate, Rp-isomer (10 µg/kg/minute; n = 8), ghrelin + microtubule polymerization inhibitor nocodazole (0.3 µg/kg/minute; n = 7), or ghrelin + actin polymerization inhibitor cytochalasin D (0.3 µg/kg/minute; n = 6). Compared with vehicle infusion, RI ghrelin induced a significant anti-natriuresis (urine Na+ excretion was reduced by 53.7% ± 6.8%; P < 0.001). This effect was abolished during concomitant PKA or microtubule inhibition (106.4% ± 9.4% and 109.7% ± 10.6% of vehicle infusion, respectively; P < 0.01 from ghrelin) but not during concomitant PI3K or actin inhibition (reduced by 48.6% ± 3.9% and 52.8% ± 12.7%, respectively; P < 0.001 and P < 0.01 from vehicle, respectively; P = not significant from ghrelin). Infusions had no effect on mean arterial pressure. Western blot analysis demonstrated that CD membrane but not total αENaC expression increased in response to ghrelin infusion compared with vehicle, (0.39 ± 0.05 vs 0.12 ± 0.02 arbitrary units; P < 0.01). This effect was abolished during PKA or microtubule inhibition but persisted during PI3K or actin inhibition. Neural precursor cell expressed, developmentally down-regulated 4 isoform 2 (Nedd4-2) dependent internalization of αENaC was not affected by ghrelin, indicating that microtubule-dependent forward trafficking of αENaC is necessary for anti-natriuretic responses to ghrelin. Taken together, these studies highlight the importance of PKA and microtubule polymerization in ghrelin-induced αENaC-mediated Na+ reabsorption.

7.
J Am Heart Assoc ; 8(9): e012016, 2019 05 07.
Article in English | MEDLINE | ID: mdl-31039659

ABSTRACT

Background Previous studies demonstrated that angiotensin (Ang) III , not Ang II , is the predominant endogenous agonist for Ang type-2 receptor ( AT 2R)-induced natriuresis in normal rats, and that hypertensive 12-week-old spontaneously hypertensive rats ( SHR ) lack natriuretic responses to Ang III . This study tested whether prehypertensive SHR already have defective Ang III -induced natriuresis and determined possible mechanisms. Methods and Results Female and male normotensive 4-week-old SHR and Wistar Kyoto rats were studied after 24-hour systemic AT 1R blockade. Left kidneys received 30 minute renal interstitial infusions of vehicle followed by Ang III (3.5, 7.0, 14, and 28 nmol/kg per min; each dose for 30 minutes). Right kidneys received vehicle infusions. In 4-week-old Wistar Kyoto rats, renal interstitial Ang III increased urine sodium (Na+) excretion but failed to induce natriuresis in 4-week-old SHR . Renal Ang III levels were similar between Wistar Kyoto rats and SHR , making increased Ang III degradation as a possible cause for defective natriuresis in SHR unlikely. In Wistar Kyoto rats, renal interstitial Ang III induced translocation of AT 2Rs to apical plasma membranes of renal proximal tubule cells. Simultaneously, Ang III induced retraction of the major Na+ transporter Na+-H+ exchanger-3 ( NHE -3) from apical membranes and internalization of Na+/K+ ATP ase ( NKA ) from basolateral membranes of renal proximal tubule cells. Consistent with NHE -3 and NKA retraction, Ang III increased pS er552- NHE -3 and decreased pS er23- NKA . In contrast, in SHR , intrarenal Ang III failed to induce AT 2R translocation, NHE -3 or NKA retraction, pS er552- NHE -3 phosphorylation, or pS er23- NKA dephosphorylation. Conclusions These results indicate impaired Ang III / AT 2R signaling as a possible primary defect in prehypertensive SHR .


Subject(s)
Angiotensin III/administration & dosage , Arterial Pressure/drug effects , Kidney/drug effects , Natriuresis/drug effects , Prehypertension/metabolism , Receptor, Angiotensin, Type 2/agonists , Renin-Angiotensin System/drug effects , Animals , Disease Models, Animal , Female , Kidney/metabolism , Kidney/physiopathology , Male , Phosphorylation , Prehypertension/physiopathology , Rats, Inbred SHR , Rats, Inbred WKY , Receptor, Angiotensin, Type 2/metabolism , Signal Transduction , Sodium-Hydrogen Exchanger 3/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism
8.
Hypertension ; 73(6): 1258-1265, 2019 06.
Article in English | MEDLINE | ID: mdl-31030607

ABSTRACT

Activation of the renal D1R (dopamine D1-like receptor) or AT2R (angiotensin II type-2 receptor), individually or both, simultaneously, is necessary in the normal regulation of renal sodium (Na+) transport and blood pressure. However, little is known regarding the precise mechanism of this interaction. Pharmacological stimulation, membrane biotinylation, and cell surface immunofluorescence were used to study the effect of the D1R/AT2R interaction in human renal proximal tubule cells. D1R activation of GαS stimulates AC (adenylyl cyclase) and induces apical plasma membrane recruitment of AT2Rs. We now show for the first time the reciprocal reaction, AT2R stimulation with Ang III (angiotensin III) leads to the apical plasma membrane recruitment of the D1R. The cell-permeable second messenger analogs of cAMP (8-Br-cAMP) or cGMP (8-Br-cGMP) induce translocation of both D1R and AT2R to the plasma membrane. Inhibition of PKA (protein kinase A) with Rp-cAMPS and PKG (protein kinase G) with Rp-8-CPT-cGMPS blocks D1R and AT2R recruitment, respectively, indicating that both PKA and PKG are necessary for D1R and AT2R trafficking. Both 8-Br-cAMP and 8-Br-cGMP activate PP2A (protein phosphatase 2A), which is necessary for both plasma membrane recruitment of D1R and AT2R and the inhibition of sodium hydrogen exchanger 3-dependent Na+ transport. These studies provide insights into the D1R/AT2R transregulation mechanisms that play a crucial role in maintaining Na+ and ultimately blood pressure homeostasis.


Subject(s)
Blood Pressure/physiology , Kidney Tubules, Proximal/metabolism , Protein Phosphatase 2/metabolism , Receptor, Angiotensin, Type 2/metabolism , Receptors, Dopamine D1/metabolism , Sodium/metabolism , Blotting, Western , Cell Membrane/metabolism , Cells, Cultured , Humans , Ion Transport , Kidney Tubules, Proximal/cytology , Natriuresis , Signal Transduction
9.
J Am Heart Assoc ; 8(4): e010926, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30764689

ABSTRACT

Background Sustained pressure overload leads to changes in cardiac metabolism, function, and structure. Both time course and causal relationships between these changes are not fully understood. Therefore, we studied spontaneously hypertensive rats (SHR) during early hypertension development and compared them to control Wistar Kyoto rats. Methods and Results We serially evaluated myocardial glucose uptake rates (Ki) with dynamic 2-[18F] fluoro-2-deoxy-D-glucose positron emission tomography, and ejection fraction and left ventricular mass to body weight ratios with cardiac magnetic resonance imaging in vivo, determined glucose uptake and oxidation rates in isolated perfused hearts, and analyzed metabolites, mammalian target of rapamycin activity and endoplasmic reticulum stress in dissected hearts. When compared with Wistar Kyoto rats, SHR demonstrated increased glucose uptake rates (Ki) in vivo, and reduced ejection fraction as early as 2 months of age when hypertension was established. Isolated perfused SHR hearts showed increased glucose uptake and oxidation rates starting at 1 month. Cardiac metabolite analysis at 2 months of age revealed elevated pyruvate, fatty acyl- and branched chain amino acid-derived carnitines, oxidative stress, and inflammation. Mammalian target of rapamycin activity increased in SHR beginning at 2 months. Left ventricular mass to body weight ratios and endoplasmic reticulum stress were elevated in 5 month-old SHR. Conclusions Thus, in a genetic hypertension model, chronic cardiac pressure overload promptly leads to increased myocardial glucose uptake and oxidation, and to metabolite abnormalities. These coincide with, or precede, cardiac dysfunction while left ventricular hypertrophy develops only later. Myocardial metabolic changes may thus serve as early diagnostic markers for hypertension-induced left ventricular hypertrophy.


Subject(s)
Blood Pressure/physiology , Heart Ventricles/physiopathology , Hypertension/metabolism , Hypertrophy, Left Ventricular/metabolism , Myocardium/pathology , Oxidative Stress , Ventricular Function, Left/physiology , Animals , Disease Models, Animal , Disease Progression , Heart Ventricles/diagnostic imaging , Heart Ventricles/metabolism , Hypertension/etiology , Hypertension/physiopathology , Hypertrophy, Left Ventricular/etiology , Hypertrophy, Left Ventricular/physiopathology , Male , Positron-Emission Tomography , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Tomography, X-Ray Computed
10.
Am J Physiol Renal Physiol ; 315(4): F1058-F1066, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29923768

ABSTRACT

The intrarenal ghrelin receptor (GR) is localized to collecting duct (CD) cells, where it increases epithelial Na+ channel (αENaC)-dependent sodium reabsorption in rodents. We hypothesized that chronic GR inhibition with intrarenal GR siRNA lowers blood pressure (BP) in angiotensin II-dependent hypertension via reductions in αENaC-dependent sodium reabsorption. Uninephrectomized Sprague-Dawley rats ( n = 121) received subcutaneous osmotic pumps for chronic systemic delivery of angiotensin II or vehicle (5% dextrose in water). Rats also received intrarenal infusion of vehicle, GR siRNA, or scrambled (SCR) siRNA. In rats receiving intrarenal vehicle or intrarenal SCR siRNA, systemic angiotensin II infusion increased sodium retention and BP on day 1, and BP remained elevated throughout the 5-day study. These rats also demonstrated increased CD GR expression after 5 days of infusion. However, intrarenal GR siRNA infusion prevented angiotensin II-mediated sodium retention on day 1, induced a continuously negative cumulative sodium balance compared with angiotensin II alone, and reduced BP chronically. Glomerular filtration rate and renal blood flow remained unchanged in GR siRNA-infused rats. Systemic angiotensin II infusion also increased serum aldosterone levels, CD αENaC, and phosphorylated serum and glucocorticoid-inducible kinase 1 expression in rats with intrarenal SCR siRNA; however, these effects were not observed in the presence of intrarenal GR siRNA, despite exposure to the same systemic angiotensin II. These data demonstrate that chronic inhibition of intrarenal GR activity significantly reduces αENaC-dependent sodium retention, resulting in a negative cumulative sodium balance, thereby ameliorating angiotensin II-induced hypertension in rats. Renal GRs represent a novel therapeutic target for the treatment of hypertension and other sodium-retaining states.


Subject(s)
Angiotensin II/metabolism , Hypertension/metabolism , Kidney/metabolism , Receptors, Ghrelin/metabolism , Angiotensin II/pharmacology , Animals , Blood Pressure/drug effects , Female , Glomerular Filtration Rate/drug effects , Rats, Sprague-Dawley , Renal Circulation/drug effects , Sodium/metabolism
11.
PLoS One ; 13(4): e0189464, 2018.
Article in English | MEDLINE | ID: mdl-29642240

ABSTRACT

RATIONALE: Salt sensitivity of blood pressure affects >30% of the hypertensive and >15% of the normotensive population. Variants of the electrogenic sodium bicarbonate cotransporter NBCe2 gene, SLC4A5, are associated with increased blood pressure in several ethnic groups. SLC4A5 variants are also highly associated with salt sensitivity, independent of hypertension. However, little is known about how NBCe2 contributes to salt sensitivity, although NBCe2 regulates renal tubular sodium bicarbonate transport. We hypothesized that SLC4A5 rs10177833 and rs7571842 increase NBCe2 expression and human renal proximal tubule cell (hRPTC) sodium transport and may be a cause of salt sensitivity of blood pressure. OBJECTIVE: To characterize the hRPTC ion transport of wild-type (WT) and homozygous variants (HV) of SLC4A5. METHODS AND RESULTS: The expressions of NBCe2 mRNA and protein were not different between hRPTCs carrying WT or HV SLC4A5 before or after dopaminergic or angiotensin (II and III) stimulation. However, luminal to basolateral sodium transport, NHE3 protein, and Cl-/HCO3- exchanger activity in hRPTCs were higher in HV than WT SLC4A5. Increasing intracellular sodium enhanced the apical location of NBCe2 in HV hRPTCs (4.24±0.35% to 11.06±1.72% (P<0.05, N = 3, 2-way ANOVA, Holm-Sidak test)) as determined by Total Internal Reflection Fluorescence Microscopy (TIRFM). In hRPTCs isolated from kidney tissue, increasing intracellular sodium enhanced bicarbonate-dependent pH recovery rate and increased NBCe2 mRNA and protein expressions to a greater extent in HV than WT SLC4A5 (+38.00±6.23% vs HV normal salt (P<0.01, N = 4, 2-way ANOVA, Holm-Sidak test)). In hRPTCs isolated from freshly voided urine, bicarbonate-dependent pH recovery was also faster in those from salt-sensitive and carriers of HV SLC4A5 than from salt-resistant and carriers of WT SLC4A5. The faster NBCe2-specific bicarbonate-dependent pH recovery rate in HV SCL4A5 was normalized by SLC4A5- but not SLC4A4-shRNA. The binding of purified hepatocyte nuclear factor type 4A (HNF4A) to DNA was increased in hRPTCs carrying HV SLC4A5 rs7571842 but not rs10177833. The faster NBCe2-specific bicarbonate-dependent pH recovery rate in HV SCL4A5 was abolished by HNF4A antagonists. CONCLUSION: NBCe2 activity is stimulated by an increase in intracellular sodium and is hyper-responsive in hRPTCs carrying HV SLC4A5 rs7571842 through an aberrant HNF4A-mediated mechanism.


Subject(s)
Bicarbonates/metabolism , Kidney Tubules, Proximal/cytology , Polymorphism, Single Nucleotide , Sodium-Bicarbonate Symporters/genetics , Sodium/metabolism , Blood Pressure/drug effects , Blood Pressure/genetics , Dopamine Agonists/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Hepatocyte Nuclear Factor 4/metabolism , Homozygote , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Protein Transport/drug effects , Protein Transport/genetics , Receptors, Dopamine/metabolism , Sodium-Bicarbonate Symporters/metabolism
12.
Circ Res ; 119(4): 532-43, 2016 Aug 05.
Article in English | MEDLINE | ID: mdl-27323774

ABSTRACT

RATIONALE: Compound 21 (C-21) is a highly selective nonpeptide angiotensin AT2 receptor (AT2R) agonist. OBJECTIVE: To test the hypothesis that chronic AT2R activation with C-21 induces natriuresis via an action at the renal proximal tubule (RPT) and lowers blood pressure (BP) in experimental angiotensin II (Ang II)-dependent hypertension. METHODS AND RESULTS: In rats, Ang II infusion increased both sodium (Na(+)) retention and BP on day 1, and BP remained elevated throughout the 7-day infusion period. Either intrarenal or systemic administration of C-21 prevented Ang II-mediated Na(+) retention on day 1, induced continuously negative cumulative Na(+) balance compared with Ang II alone, and reduced BP chronically. The effects of C-21 are likely to be mediated by action on the RPT as acute systemic C-21-induced natriuresis was additive to that induced by chlorothiazide and amiloride. At 24 hours of Ang II infusion, AT2R activation with C-21, both intrarenally and systemically, translocated AT2Rs from intracellular sites to the apical plasma membranes of RPT cells without altering the total cellular pool of AT2Rs and internalized/inactivated major RPT Na(+) transporters Na(+)-H(+)-exchanger-3 and Na(+)/K(+)ATPase. C-21 lowered BP to a similar degree whether administered before or subsequent to the establishment of Ang II-dependent hypertension. CONCLUSIONS: Chronic AT2R activation initiates and sustains receptor translocation to RPT apical plasma membranes, internalizes/inactivates Na(+)-H(+)-exchanger-3 and Na(+)/K(+)ATPase, prevents Na(+) retention resulting in negative cumulative Na(+) balance, and lowers BP in experimental Ang II-induced hypertension. Acting uniquely at the RPT, C-21 is a promising candidate for the treatment of hypertension and Na(+)-retaining states in humans.


Subject(s)
Angiotensin II/toxicity , Blood Pressure/physiology , Hypertension/metabolism , Natriuresis/physiology , Receptor, Angiotensin, Type 2/metabolism , Sodium/urine , Angiotensin II Type 2 Receptor Blockers/pharmacology , Animals , Blood Pressure/drug effects , Female , Hypertension/chemically induced , Hypertension/drug therapy , Natriuresis/drug effects , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 2/agonists , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Thiophenes/pharmacology , Thiophenes/therapeutic use
13.
Am J Physiol Regul Integr Comp Physiol ; 309(11): R1447-59, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26447209

ABSTRACT

The electrogenic sodium bicarbonate cotransporter (NBCe2) is encoded by SLC4A5, variants of which have been associated with salt sensitivity of blood pressure, which affects 25% of the adult population. NBCe2 is thought to mediate sodium bicarbonate cotransport primarily in the renal collecting duct, but NBCe2 mRNA is also found in the rodent renal proximal tubule (RPT). The protein expression or function of NBCe2 has not been demonstrated in the human RPT. We validated an NBCe2 antibody by shRNA and Western blot analysis, as well as overexpression of an epitope-tagged NBCe2 construct in both RPT cells (RPTCs) and human embryonic kidney 293 (HEK293) cells. Using this validated NBCe2 antibody, we found NBCe2 protein expression in the RPT of fresh and frozen human kidney slices, RPTCs isolated from human urine, and isolated RPTC apical membrane. Under basal conditions, NBCe2 was primarily found in the Golgi, while NBCe1 was primarily found at the basolateral membrane. Following an acute short-term increase in intracellular sodium, NBCe2 expression was increased at the apical membrane in cultured slices of human kidney and polarized, immortalized RPTCs. Sodium bicarbonate transport was increased by monensin and overexpression of NBCe2, decreased by NBCe2 shRNA, but not by NBCe1 shRNA, and blocked by 2,2'-(1,2-ethenediyl)bis[5-isothiocyanato-benzenesulfonic acid]. NBCe2 could be important in apical sodium and bicarbonate cotransport under high-salt conditions; the implication of the ex vivo studies to the in vivo situation when salt intake is increased remains unclear. Therefore, future studies will examine the role of NBCe2 in mediating increased renal sodium transport in humans whose blood pressures are elevated by an increase in sodium intake.


Subject(s)
Bicarbonates/metabolism , Kidney Tubules, Proximal/metabolism , Sodium-Bicarbonate Symporters/metabolism , Sodium/metabolism , Cell Membrane/metabolism , Golgi Apparatus/metabolism , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Kidney Tubules, Proximal/drug effects , Monensin/pharmacology , Protein Transport , RNA Interference , Sodium-Bicarbonate Symporters/drug effects , Sodium-Bicarbonate Symporters/genetics , Time Factors , Transfection
14.
Circ Res ; 115(3): 388-99, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24903104

ABSTRACT

RATIONALE: Compound 21 (C-21) is a highly selective nonpeptide AT2 receptor (AT2R) agonist. OBJECTIVE: To test the hypothesis that renal proximal tubule AT2Rs induce natriuresis and lower blood pressure in Sprague-Dawley rats and mice. METHODS AND RESULTS: In rats, AT2R activation with intravenous C-21 increased urinary sodium excretion by 10-fold (P<0.0001); this natriuresis was abolished by direct renal interstitial infusion of specific AT2R antagonist PD-123319. C-21 increased fractional excretion of Na(+) (P<0.05) and lithium (P<0.01) without altering renal hemodynamic function. AT2R activation increased renal proximal tubule cell apical membrane AT2R protein (P<0.001) without changing total AT2R expression and internalized/inactivated Na(+)-H(+) exchanger-3 and Na(+)/K(+)ATPase. C-21-induced natriuresis was accompanied by an increase in renal interstitial cGMP (P<0.01); C-21-induced increases in urinary sodium excretion and renal interstitial cGMP were abolished by renal interstitial nitric oxide synthase inhibitor l-N(6)-nitroarginine methyl ester or bradykinin B2 receptor antagonist icatibant. Renal AT2R activation with C-21 prevented Na(+) retention and lowered blood pressure in the angiotensin II infusion model of experimental hypertension. CONCLUSIONS: AT2R activation initiates its translocation to the renal proximal tubule cell apical membrane and the internalization of Na(+)-H(+) exchanger-3 and Na(+)/K(+)ATPase, inducing natriuresis in a bradykinin-nitric oxide-cGMP-dependent manner. Intrarenal AT2R activation prevents Na(+) retention and lowers blood pressure in angiotensin II-dependent hypertension. AT2R activation holds promise as a renal proximal tubule natriuretic/diuretic target for the treatment of fluid-retaining states and hypertension.


Subject(s)
Blood Pressure/drug effects , Hypertension, Renal/drug therapy , Natriuresis/drug effects , Receptor, Angiotensin, Type 2/agonists , Sulfonamides/pharmacology , Thiophenes/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II Type 2 Receptor Blockers/pharmacology , Animals , Blood Pressure/physiology , Bradykinin/analogs & derivatives , Bradykinin/pharmacology , Bradykinin Receptor Antagonists , Enzyme Inhibitors/pharmacology , Female , Glomerular Filtration Rate/drug effects , Glomerular Filtration Rate/physiology , Hypertension, Renal/physiopathology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/physiology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NG-Nitroarginine Methyl Ester/pharmacology , Natriuresis/physiology , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 2/genetics , Renal Circulation/drug effects , Renal Circulation/physiology
15.
Endocrinology ; 155(7): 2658-66, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24797629

ABSTRACT

Excess weight gain contributes up to 65% of the risk of primary hypertension, and the increase in blood pressure in response to high-fat diet (HFD) is preceded by significant increases in renal tubular sodium (Na(+)) reabsorption. In normal rats, intrarenal ghrelin infusion increases distal nephron-dependent Na(+) reabsorption via activation of the intrarenal ghrelin receptor (GHSR). This study focusses on the role of intrarenal GHSR-mediated Na(+) reabsorption in HFD-induced hypertension. Dahl salt-sensitive rats received standard diet or HFD for 6 weeks. Rats underwent uninephrectomy and osmotic minipump implantation for chronic intrarenal delivery of vehicle (0.25 µL/h × 28 d), selective GHSR antagonist [D-Lys-3]-growth hormone releasing peptide-6 (0.2µM/d), or GHSR inverse agonist [D-Arg(1), D-Phe(5), D-Trp(7,9), Leu(11)]-substance P (SUB-P) (3.6µM/d). HFD rats with vehicle pumps had significantly increased renal GHSR expression compared with standard diet (0.092 ± 0.005 vs 0.065 ± 0.004 arbitrary units; P < .05), whereas acyl ghrelin levels were similar (16.3±6.2 vs 15.7±8.7 pg/g tissue). HFD rats with vehicle pumps became hypertensive after 2 weeks (P < .05) and showed a significant reduction in 24-hour urine Na(+) before hypertension. At this time, these rats showed an increase in collecting duct α-epithelial Na(+) channel, thereby providing a potential mechanism for the excess Na(+) reabsorption. In contrast, HFD rats with [D-Lys-3]-growth hormone releasing peptide-6 or SUB-P pumps never became hypertensive and did not show the reduction in urine Na(+). Because SUB-P blocks the constitutive, but not ghrelin-dependent, activity of the GHSR, and HFD-induced α-epithelial Na(+) channel up-regulation was abolished during GHSR antagonism, these data suggest that HFD increases the constitutive activity of renal GHSR to increase Na(+) reabsorption and induce hypertension in rats.


Subject(s)
Diet, High-Fat/adverse effects , Hypertension/prevention & control , Kidney/drug effects , Receptors, Ghrelin/antagonists & inhibitors , Animals , Blotting, Western , Body Weight/drug effects , Eating/drug effects , Ghrelin/metabolism , Hypertension/etiology , Hypertension/metabolism , Kidney/metabolism , Kidney/surgery , Male , Nephrectomy , Oligopeptides/pharmacology , Rats , Rats, Inbred Dahl , Rats, Sprague-Dawley , Receptors, Ghrelin/metabolism , Sodium/metabolism , Sodium/pharmacokinetics , Sodium/urine , Substance P/analogs & derivatives , Substance P/pharmacology , Time Factors
16.
Kidney Int ; 84(3): 501-8, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23698230

ABSTRACT

The main distal nephron segment sodium transporters are the distal tubule chlorothiazide-sensitive sodium chloride cotransporter (NCC) and the collecting duct amiloride-sensitive epithelial sodium channel (ENaC). The infusion of ghrelin into the renal interstitium stimulates distal nephron-dependent sodium reabsorption in normal rats, but the mechanism is unknown. Here we localize renal ghrelin receptors (GR) to the cortical collecting duct (CCD). Ghrelin significantly increased phosphorylated serum/glucocorticoid-regulated kinase-1 (pSGK1), a major upstream signaling intermediate regulating ENaC. To test whether increased apical membrane αENaC induced the antinatriuresis, ghrelin was infused in the presence of acute and chronic amiloride, a selective inhibitor of ENaC. In the presence of amiloride, renal interstitial ghrelin failed to reduce urine sodium excretion, suggesting that ghrelin-induced sodium reabsorption is dependent on intact ENaC activity. While the main sodium transporter of the CCD is ENaC, NCC is also present. In response to renal interstitial ghrelin infusion, neither total nor phosphorylated NCC levels are altered. Ghrelin-induced sodium reabsorption persisted in the presence of chlorothiazide (selective inhibitor of NCC), indicating that intact NCC activity is not necessary for ghrelin-induced antinatriuresis. Finally, renal interstitial ghrelin infusion significantly increased interstitial cAMP levels and adenylyl cyclase blockade abolished ghrelin-induced antinatriuresis. Thus, GRs expressed in the CCD regulate sodium reabsorption by cAMP-induced trafficking of ENaC to the apical membrane.


Subject(s)
Cyclic AMP/physiology , Epithelial Sodium Channels/physiology , Kidney Tubules, Collecting/physiology , Receptors, Ghrelin/physiology , Signal Transduction/physiology , Sodium/metabolism , Amiloride/pharmacology , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Chlorothiazide/pharmacology , Female , Ghrelin/pharmacology , Immediate-Early Proteins/physiology , Models, Animal , Protein Serine-Threonine Kinases/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
17.
Hypertension ; 60(2): 387-95, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22689743

ABSTRACT

In angiotensin type 1 receptor-blocked rats, renal interstitial (RI) administration of des-aspartyl(1)-angiotensin II (Ang III) but not angiotensin II induces natriuresis via activation of angiotensin type 2 receptors. In the present study, renal function was documented during systemic angiotensin type 1 receptor blockade with candesartan in Sprague-Dawley rats receiving unilateral RI infusion of Ang III. Ang III increased urine sodium excretion, fractional sodium, and lithium excretion. RI coinfusion of specific angiotensin type 2 receptor antagonist PD-123319 abolished Ang III-induced natriuresis. The natriuretic response observed with RI Ang III was not reproducible with RI angiotensin (1-7) alone or together with angiotensin-converting enzyme inhibition. Similarly, neither RI angiotensin II alone or in the presence of aminopeptidase A inhibitor increased urine sodium excretion. In the absence of systemic angiotensin type 1 receptor blockade, Ang III alone did not increase urine sodium excretion, but natriuresis was enabled by the coinfusion of aminopeptidase N inhibitor and subsequently blocked by PD-123319. In angiotensin type 1 receptor-blocked rats, RI administration of aminopeptidase N inhibitor alone also induced natriuresis that was abolished by PD-123319. Ang III-induced natriuresis was accompanied by increased RI cGMP levels and was abolished by inhibition of soluble guanylyl cyclase. RI and renal tissue Ang III levels increased in response to Ang III infusion and were augmented by aminopeptidase N inhibition. These data demonstrate that endogenous intrarenal Ang III but not angiotensin II or angiotensin (1-7) induces natriuresis via activation of angiotensin type 2 receptors in the proximal tubule via a cGMP-dependent mechanism and suggest aminopeptidase N inhibition as a potential therapeutic target in hypertension.


Subject(s)
Angiotensin III/pharmacology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Receptor, Angiotensin, Type 2/metabolism , Angiotensin I/pharmacology , Angiotensin II/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II Type 2 Receptor Blockers/pharmacology , Animals , Benzimidazoles/pharmacology , Biphenyl Compounds , Female , Imidazoles/pharmacology , Models, Animal , Natriuresis/drug effects , Natriuresis/physiology , Peptide Fragments/pharmacology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 2/drug effects , Tetrazoles/pharmacology
18.
J Cardiovasc Pharmacol ; 59(4): 363-8, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22157261

ABSTRACT

Cardiac overexpression of the angiotensin II type 2 receptor (AT2 R) attenuates left ventricular (LV) remodeling after myocardial infarction (MI) in transgenic mice. We hypothesized that a novel nonpeptide AT2 R agonist, compound 21 (C21), would attenuate post-MI LV remodeling. Fifty-nine mice were studied for 28 days after 1-hour surgical occlusion-reperfusion of the left anterior descending coronary artery. Immediately thereafter, 23 mice received 0.3 mg·kg·d of C21 via Alzet osmotic minipump, 16 received 10 mg·kg·d of the AT1 R antagonist candesartan in drinking water, and 20 were untreated controls. Cardiac magnetic resonance imaging measured ejection fraction (EF), LV end-systolic, and end-diastolic volumes (ESVI and EDVI) indexed to weight serially post MI. Infarct size was measured on day 1 by late gadolinium-enhanced cardiac magnetic resonance imaging. At baseline, heart rate, blood pressure, EDVI, ESVI, and EF were similar between groups. Mean infarct size (42%-45% of LV mass) was similar between groups. C21-treated animals demonstrated adverse LV remodeling (increased EDVI and ESVI at all post-MI time points) compared with control. Candesartan therapy preserved left ventricular EF at day 28 compared with the C21-treated group. Thus, direct stimulation of the AT2 R by C21 at 0.3 mg·kg·d does not attenuate post-MI LV remodeling in reperfused MI in mice.


Subject(s)
Myocardial Infarction/drug therapy , Receptor, Angiotensin, Type 2/agonists , Ventricular Remodeling/drug effects , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Benzimidazoles/pharmacology , Biphenyl Compounds , Coronary Occlusion/complications , Disease Models, Animal , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/physiopathology , Tetrazoles/pharmacology , Time Factors
19.
Hypertension ; 59(2): 437-45, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22203736

ABSTRACT

Renal dopamine D(1)-like receptors (D(1)Rs) and angiotensin type 2 receptors (AT(2)Rs) are important natriuretic receptors counterbalancing angiotensin type 1 receptor-mediated tubular sodium reabsorption. Here we explore the mechanisms of D(1)R and AT(2)R interactions in natriuresis. In uninephrectomized, sodium-loaded Sprague-Dawley rats, direct renal interstitial infusion of the highly selective D(1)R agonist fenoldopam induced a natriuretic response that was abolished by the AT(2)R-specific antagonist PD-123319 or by microtubule polymerization inhibitor nocodazole but not by actin polymerization inhibitor cytochalasin D. By confocal microscopy and immunoelectron microscopy, fenoldopam translocated AT(2)Rs from intracellular sites to the apical plasma membranes of renal proximal tubule cells, and this translocation was abolished by nocodazole. Because D(1)R activation induces natriuresis via an adenylyl cyclase/cAMP signaling pathway, we explored whether this pathway is responsible for AT(2)R recruitment and AT(2)R-mediated natriuresis. Renal interstitial coinfusion of the adenylyl cyclase activator forskolin and 3-isobutly-1-methylxanthine induced natriuresis that was abolished either by PD-123319 or nocodazole but was unaffected by specific the D(1)R antagonist SCH-23390. Coadministration of forskolin and 3-isobutly-1-methylxanthine also translocated AT(2)Rs to the apical plasma membranes of renal proximal tubule cells; this translocation was abolished by nocodazole but was unaffected by SCH-23390. The results demonstrate that D(1)R-induced natriuresis requires AT(2)R recruitment to the apical plasma membranes of renal proximal tubule cells in a microtubule-dependent manner involving an adenylyl cyclase/cAMP signaling pathway. These studies provide novel insights regarding the mechanisms whereby renal D(1)Rs and AT(2)Rs act in concert to promote sodium excretion in vivo.


Subject(s)
Kidney Tubules, Proximal/physiology , Microtubules/physiology , Natriuresis/physiology , Receptor, Angiotensin, Type 2/physiology , Receptors, Dopamine D1/physiology , Signal Transduction/physiology , Angiotensin II Type 2 Receptor Blockers/pharmacology , Animals , Cyclic AMP/physiology , Female , Fenoldopam/pharmacology , Imidazoles/pharmacology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/ultrastructure , Microtubules/drug effects , Microtubules/ultrastructure , Models, Animal , Natriuresis/drug effects , Nocodazole/pharmacology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 2/drug effects , Receptors, Dopamine D1/agonists , Receptors, Dopamine D1/drug effects , Signal Transduction/drug effects , Sodium, Dietary/pharmacology , Tubulin Modulators/pharmacology
20.
Hypertension ; 58(1): 107-13, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21482955

ABSTRACT

cGMP functions as an extracellular (paracrine) messenger acting at the renal proximal tubule and is an important modulator of pressure-natriuresis (P-N). The signaling pathway activated by cGMP in the tubule cell basolateral membrane remains unknown. We hypothesized that renal interstitial microinfusion of cGMP (50 nmol/kg per minute) or P-N would be accompanied by increased renal protein levels of phospho-Src (Tyr 416) and that the natriuresis would be decreased by Src inhibition. Renal interstitial cGMP-induced natriuresis was blocked by Src inhibitor PP2 (2.0±0.4 versus 0.5±0.01 µEq/g per minute; P<0.001). The inactive analog of PP2, PP3, had no effect on cGMP-induced natriuresis. SU6656, another Src inhibitor, also inhibited cGMP-induced natriuresis (2.0±0.4 versus 1.02±0.01 µEq/g per minute; P<0.001). Renal interstitial cGMP infusion increased phospho-Src protein levels 5.6-fold at 15 minutes and 6.8-fold at 30 minutes compared with vehicle infusion but returned toward basal levels after 60 minutes. PP2 also blunted P-N (3.1±0.1 versus 1.1±0.3 µEq/g per minute; P<0.01) despite a similar increase in blood pressure. PP3 had no effect on P-N. Phospho-Src protein levels increased during P-N in vehicle- (1.8-fold) and PP3-treated (2.1-fold) groups compared with the sham-operated group. PP2 blocked the pressure-induced increase in renal phospho-Src protein levels. PP2 had no effect on renal hemodynamics but decreased both fractional excretion of Na(+) and lithium. Both extracellular cGMP and increased renal perfusion pressure increased renal phospho-Src protein levels and induced natriuresis in an Src-dependent manner, demonstrating that Src is an important downstream signaling molecule for extracellular cGMP-induced natriuresis.


Subject(s)
Cyclic GMP/analogs & derivatives , Extracellular Fluid/metabolism , Hypertension/metabolism , Kidney/metabolism , Natriuresis/drug effects , Sodium/urine , Thionucleotides/pharmacology , src-Family Kinases/metabolism , Animals , Blood Pressure/physiology , Cyclic GMP/pharmacology , Disease Models, Animal , Female , Hypertension/physiopathology , Kidney/drug effects , Natriuresis/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...