Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 128
Filter
1.
J Clin Hypertens (Greenwich) ; 23(5): 1008-1016, 2021 05.
Article in English | MEDLINE | ID: mdl-33694311

ABSTRACT

Angiotensin-converting enzyme inhibitors (ACEi) are part of the indicated treatment in hypertensive African Americans. ACEi have blood pressure-independent effects that may make them preferred for certain patients. We aimed to evaluate the impact of ACEi on anti-fibrotic biomarkers in African American hypertensive patients with left ventricular hypertrophy (LVH). We conducted a post hoc analysis of a randomized controlled trial in which hypertensive African American patients with LVH and vitamin D deficiency were randomized to receive intensive antihypertensive therapy plus vitamin D supplementation or placebo. We selected patients who had detectable lisinopril (lisinopril group) in plasma using liquid-chromatography/mass spectrometry analysis and compared them to subjects who did not (comparison group) at the one-year follow-up. The pro-fibrotic marker type 1 procollagen C-terminal propeptide (PICP) and the anti-fibrotic markers matrix metalloproteinase-1 (MMP-1), tissue inhibitor of metalloproteinases 1 (TIMP-1), telopeptide of collagen type I (CITP), and N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP) peptide were measured. Sixty-six patients were included, and the mean age was 46.2 ± 8 years. No difference was observed in the number and intensity of antihypertensive medications prescribed in each group. Patients with detectable lisinopril had lower blood pressure than those in the comparison group. The anti-fibrotic markers Ac-SDKP, MMP-1, and MMP-1/TIMP-1 ratio were higher in patients with detectable ACEi (all p < .05). In a model adjusted for systolic blood pressure, MMP-1/TIMP-1 (p = .02) and Ac-SDKP (p < .001) levels were associated with lisinopril. We conclude that ACEi increase anti-fibrotic biomarkers in hypertensive African Americans with LVH, suggesting that they may offer added benefit over other agents in such patients.


Subject(s)
Black or African American , Hypertension , Adult , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Biomarkers , Humans , Hypertension/drug therapy , Hypertrophy, Left Ventricular/drug therapy , Middle Aged
2.
Cardiovasc Res ; 116(12): 1995-2008, 2020 10 01.
Article in English | MEDLINE | ID: mdl-31825460

ABSTRACT

AIMS: Activation of the angiotensin (Ang)-(1-7)/Mas receptor (R) axis protects from sympathetic overactivity. Endocytic trafficking is an essential process that regulates receptor (R) function and its ultimate cellular responses. We investigated whether the blunted responses to Ang-(1-7) in hypertensive rats are associated to an alteration in MasR trafficking. METHODS AND RESULTS: Brainstem neurons from Wistar-Kyoto (WKY) or spontaneously hypertensive rats (SHRs) were investigated for (i) Ang-(1-7) levels and binding and MasR expression, (ii) Ang-(1-7) responses (arachidonic acid and nitric oxide release and Akt and ERK1/2 phosphorylation), and (iii) MasR trafficking. Ang-(1-7) was determined by radioimmunoassay. MasR expression and functionality were evaluated by western blot and binding assays. MasR trafficking was evaluated by immunofluorescence. Ang-(1-7) treatment induced an increase in nitric oxide and arachidonic acid release and ERK1/2 and Akt phosphorylation in WKY neurons but did not have an effect in SHR neurons. Although SHR neurons showed greater MasR expression, Ang-(1-7)-elicited responses were substantially diminished presumably due to decreased Ang-(1-7) endogenous levels concomitant with impaired binding to its receptor. Through immunocolocalization studies, we evidenced that upon Ang-(1-7) stimulation MasRs were internalized through clathrin-coated pits and caveolae into early endosomes and slowly recycled back to the plasma membrane. However, the fraction of internalized MasRs into early endosomes was larger and the fraction of MasRs recycled back to the plasma membrane was smaller in SHR than in WKY neurons. Surprisingly, in SHR neurons but not in WKY neurons, Ang-(1-7) induced MasR translocation to the nucleus. Nuclear MasR expression and Ang-(1-7) levels were significantly greater in the nuclei of Ang-(1-7)-stimulated SHR neurons, indicating that the MasR is translocated with its ligand bound to it. CONCLUSION: MasRs display differential trafficking in brainstem neurons from SHRs, which may contribute to the impaired responses to Ang-(1-7).


Subject(s)
Angiotensin I/pharmacology , Brain Stem/drug effects , Hypertension/metabolism , Neurons/drug effects , Peptide Fragments/pharmacology , Proto-Oncogene Proteins/agonists , Receptors, G-Protein-Coupled/agonists , Active Transport, Cell Nucleus , Animals , Animals, Newborn , Arachidonic Acid/metabolism , Brain Stem/metabolism , Brain Stem/physiopathology , Cells, Cultured , Disease Models, Animal , Endocytosis , Extracellular Signal-Regulated MAP Kinases/metabolism , Hypertension/physiopathology , Ligands , Neurons/metabolism , Nitric Oxide/metabolism , Phosphorylation , Proto-Oncogene Mas , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats, Inbred SHR , Rats, Inbred WKY , Receptors, G-Protein-Coupled/metabolism
3.
Am J Physiol Renal Physiol ; 316(6): F1218-F1226, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30838873

ABSTRACT

Afferent arteriole (Af-Art) diameter regulates pressure and flow into the glomerulus, which are the main determinants of the glomerular filtration rate. Thus, Af-Art resistance is crucial for Na+ filtration. Af-Arts play a role as integrative centers, where systemic and local systems interact to determine the final degree of resistance. The tubule of a single nephron contacts an Af-Art of the same nephron at two locations: in the transition of the thick ascending limb to the distal tubule (macula densa) and again in the connecting tubule. These two sites are the anatomic basis of two intrinsic feedback mechanisms: tubule-glomerular feedback and connecting tubule-glomerular feedback. The cross communications between the tubules and Af-Arts integrate tubular Na+ and water processing with the hemodynamic conditions of the kidneys. Tubule-glomerular feedback provides negative feedback that tends to avoid salt loss, and connecting tubule-glomerular feedback provides positive feedback that favors salt excretion by modulating tubule-glomerular feedback (resetting it) and increasing glomerular filtration rate. These feedback mechanisms are also exposed to systemic modulators (hormones and the nervous system); however, they can work in isolated kidneys or nephrons. The exaggerated activation or absence of any of these mechanisms may lead to disequilibrium in salt and water homeostasis, especially in extreme conditions (e.g., high-salt diet/low-salt diet) and may be part of the pathogenesis of some diseases. In this review, we focus on molecular signaling, feedback interactions, and the physiological roles of these two feedback mechanisms.


Subject(s)
Epithelial Sodium Channels/metabolism , Glomerular Filtration Rate , Kidney Glomerulus/blood supply , Kidney Tubules/metabolism , Renal Circulation , Sodium/metabolism , Water-Electrolyte Balance , Animals , Feedback, Physiological , Hemodynamics , Humans , Kidney Tubules/physiopathology , Water-Electrolyte Imbalance/metabolism , Water-Electrolyte Imbalance/physiopathology
4.
Curr Hypertens Rep ; 21(1): 8, 2019 01 18.
Article in English | MEDLINE | ID: mdl-30659366

ABSTRACT

PURPOSE OF REVIEW: In this review, we summarized the current knowledge of connecting tubule-glomerular feedback (CTGF), a novel mechanism of renal microcirculation regulation that integrates sodium handling in the connecting tubule (CNT) with kidney hemodynamics. RECENT FINDINGS: Connecting tubule-glomerular feedback is a crosstalk communication between the CNT and the afferent arteriole (Af-Art), initiated by sodium chloride through the epithelial sodium channel (ENaC). High sodium in the CNT induces Af-Art vasodilation, increasing glomerular pressure and the glomerular filtration rate and favoring sodium excretion. CTGF antagonized and reset tubuloglomerular feedback and thus increased sodium excretion. CTGF is absent in spontaneous hypertensive rats and is overactivated in Dahl salt-sensitive rats. CTGF is also modulated by angiotensin II and aldosterone. CTGF is a feedback mechanism that integrates sodium handling in the CNT with glomerular hemodynamics. Lack of CTGF could promote hypertension, and CTGF overactivation may favor glomerular damage and proteinuria. More studies are needed to explore the alterations in renal microcirculation and the role of these alterations in the genesis of hypertension and glomerular damage in animals and humans. KEY POINTS: • CTGF is a vasodilator mechanism that regulates afferent arteriole resistance. • CTGF is absent in spontaneous hypertensive rats and overactivated in Dahl salt-sensitive rats. • CTGF in excess may promote glomerular damage and proteinuria, while the absence may participate in sodium retention and hypertension.


Subject(s)
Kidney Glomerulus/physiology , Kidney Tubules/physiology , Microcirculation/physiology , Renal Circulation/physiology , Animals , Feedback , Humans , Hypertension/physiopathology , Proteinuria/physiopathology , Rats , Sodium/metabolism
5.
Am J Physiol Renal Physiol ; 316(1): F195-F203, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30403163

ABSTRACT

The antifibrotic peptide N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP) is released from thymosin-ß4 (Tß4) by the meprin-α and prolyl oligopeptidase (POP) enzymes and is hydrolyzed by angiotensin-converting enzyme (ACE). Ac-SDKP is present in urine; however, it is not clear whether de novo tubular release occurs or if glomerular filtration is the main source. We hypothesized that Ac-SDKP is released into the lumen of the nephrons and that it exerts an antifibrotic effect. We determined the presence of Tß4, meprin-α, and POP in the kidneys of Sprague-Dawley rats. The stop-flow technique was used to evaluate Ac-SDKP formation in different nephron segments. Finally, we decreased Ac-SDKP formation by inhibiting the POP enzyme and evaluated the long-term effect in renal fibrosis. The Tß4 precursor and the releasing enzymes meprin-α and POP were expressed in the kidneys. POP enzyme activity was almost double that in the renal medulla compared with the renal cortex. With the use of the stop-flow technique, we detected the highest Ac-SDKP concentrations in the distal nephron. The infusion of a POP inhibitor into the kidney decreased the amount of Ac-SDKP in distal nephron segments and in the proximal nephron to a minor extent. An ACE inhibitor increased the Ac-SDKP content in all nephron segments, but the increase was highest in the distal portion. The chronic infusion of a POP inhibitor increased kidney medullary fibrosis, which was prevented by Ac-SDKP. We conclude that Ac-SDKP is released by the nephron and is part of an important antifibrotic system in the kidney.


Subject(s)
Kidney Diseases/metabolism , Kidney Medulla/metabolism , Nephrons/metabolism , Oligopeptides/metabolism , Animals , Disease Models, Animal , Fibrosis , Kidney Diseases/pathology , Kidney Diseases/prevention & control , Kidney Medulla/pathology , Male , Metalloendopeptidases/metabolism , Prolyl Oligopeptidases , Rats, Sprague-Dawley , Serine Endopeptidases/metabolism , Signal Transduction , Thymosin/metabolism
6.
Am J Physiol Renal Physiol ; 315(6): F1708-F1713, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30303713

ABSTRACT

Zucker obese rats (ZOR) have higher glomerular capillary pressure (PGC) that can cause renal damage. PGC is controlled by afferent (Af-Art) and efferent arteriole (Ef-Art) resistance. Af-Art resistance is regulated by factors that regulate other arterioles, such as myogenic response. In addition, it is also regulated by 2 intrinsic feedback mechanisms: 1) tubuloglomerular feedback (TGF) that causes Af-Art constriction in response to increased NaCl in the macula densa and 2) connecting tubule glomerular feedback (CTGF) that causes Af-Art dilatation in response to an increase in NaCl transport in the connecting tubule via the epithelial sodium channel. Since CTGF is an Af-Art dilatory mechanism, we hypothesized that increased CTGF contributes to TGF attenuation, which in turn increases PGC in ZOR. We performed a renal micropuncture experiment and measured stop-flow pressure (PSF), which is an indirect measurement of PGC in ZOR. Maximal TGF response at 40 nl/min was attenuated in ZOR (4.47 ± 0.60 mmHg) in comparison to the Zucker lean rats (ZLR; 8.54 ± 0.73 mmHg, P < 0.05), and CTGF was elevated in ZOR (5.34 ± 0.87 mmHg) compared with ZLR (1.12 ± 1.28 mmHg, P < 0.05). CTGF inhibition with epithelial sodium channel blocker normalized the maximum PSF change in ZOR indicating that CTGF plays a significant role in TGF attenuation (ZOR, 10.67 ± 1.07 mmHg vs. ZLR, 9.5 ± 1.53 mmHg). We conclude that enhanced CTGF contributes to TGF attenuation in ZOR and potentially contribute to progressive renal damage.


Subject(s)
Connective Tissue Growth Factor/metabolism , Kidney Diseases/etiology , Kidney Glomerulus/blood supply , Kidney Tubules/metabolism , Microcirculation , Obesity/complications , Proteinuria/etiology , Renal Circulation , Animals , Arterial Pressure , Disease Models, Animal , Feedback, Physiological , Glomerular Filtration Rate , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Diseases/physiopathology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Kidney Tubules/pathology , Kidney Tubules/physiopathology , Proteinuria/metabolism , Proteinuria/pathology , Proteinuria/physiopathology , Rats, Zucker , Signal Transduction , Up-Regulation
7.
Am J Hypertens ; 31(8): 902-909, 2018 07 16.
Article in English | MEDLINE | ID: mdl-29722788

ABSTRACT

BACKGROUND: Obesity is a public health problem, associated with salt sensitive hypertension, kidney inflammation, and fibrosis. N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP) is a tetra peptide with anti-inflammatory and anti-fibrotic properties. However, its effect on preventing kidney damage in obesity is unknown. We hypothesized that Zucker obese (ZO) rats on a high-salt (HS) diet develop renal damage, inflammation, fibrosis, and this is prevented with Ac-SDKP treatment. METHODS: Zucker lean (ZL) and ZO rats (8 weeks old) were treated with Ac-SDKP (1.6 mg/kg/day) while maintained on either a normal-salt (NS; 0.4%) or HS (4%) diet for 8 weeks. Systolic blood pressure (SBP), albuminuria, renal inflammation, and fibrosis were evaluated. RESULTS: HS diet increased macrophage infiltration in the kidneys of both ZL and ZO rats but was significantly higher in ZO rats receiving the HS diet (ZL + NS, 13.9 ± 1.3 vs. ZL + HS, 19.14 ± 1.5 and ZO + NS, 25.5 ± 1.4 vs. ZO + HS, 87.8 ± 10.8 cells/mm2; P < 0.05). Ac-SDKP prevented macrophage infiltration in ZO rats (ZO + HS + Ac-SDKP, 32.18 ± 2.4 cells/mm2; P < 0.05). Similarly, glomerulosclerosis, cortical, and medullary interstitial fibrosis were increased in ZO rats fed the HS diet, and Ac-SDKP attenuated these alterations (P < 0.05). SBP was increased in ZO rats fed the HS diet (ZO + NS, 121.3 ± 8.9 vs. ZO + HS, 164 ± 6.9 mm Hg; P < 0.05), and it was significantly decreased with Ac-SDKP treatment (ZO + HS + Ac-SDKP, 144.05 ± 14.1 mm Hg; P = 0.004). Albuminuria was higher in ZO rats than in ZL rats; however, neither HS nor Ac-SDKP treatment affected it. CONCLUSIONS: Ac-SDKP treatment in ZO rats fed a HS diet prevented renal damage by reducing inflammation, fibrosis, and SBP.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antihypertensive Agents/pharmacology , Glomerulonephritis/prevention & control , Hypertension/prevention & control , Kidney/drug effects , Obesity/drug therapy , Oligopeptides/pharmacology , Sodium Chloride, Dietary , Albuminuria/etiology , Albuminuria/physiopathology , Albuminuria/prevention & control , Animals , Blood Pressure/drug effects , Disease Models, Animal , Fibrosis , Glomerulonephritis/etiology , Glomerulonephritis/pathology , Glomerulonephritis/physiopathology , Hypertension/etiology , Hypertension/physiopathology , Kidney/pathology , Macrophages/drug effects , Macrophages/pathology , Male , Obesity/complications , Obesity/physiopathology , Rats, Zucker
8.
Hypertension ; 71(6): 1133-1142, 2018 06.
Article in English | MEDLINE | ID: mdl-29632102

ABSTRACT

Thymosin ß4 (Tß4), a ubiquitous peptide, regulates several cellular processes that include cell morphology, wound healing, and inflammatory response. Administration of exogenous Tß4 is protective in diabetic nephropathy and in a unilateral ureteral obstruction model. However, the role of endogenous Tß4 in health and disease conditions remains unclear. To elucidate the pathophysiological role of endogenous Tß4 in hypertension, we examined angiotensin-II (Ang-II)-induced renal and cardiac damage in Tß4 knockout (Tß4 KO) mice. Tß4 KO and wild-type C57BL/6 mice were infused continuously for 6 weeks with either vehicle or Ang-II (980 ng/kg per minute). At baseline, Tß4 deficiency did not affect renal and cardiac function. Systolic blood pressure in the Ang-II group was similar in wild-type and Tß4 KO mice (wild-type Ang-II, 179.25±10.11 mm Hg; Tß4 KO Ang-II, 169.81±6.54 mm Hg). Despite the similar systolic blood pressure after Ang-II infusion, Tß4-deficient mice had dramatically increased albuminuria and decreased nephrin expression in the kidney (P<0.005). In the heart of Tß4 KO mice, Ang-II reduced ejection fraction and shortening fraction (ejection fraction: wild-type Ang-II 77.95%±1.03%; Tß4 KO Ang-II 62.58%±3.25%; P<0.005), which was accompanied by cardiac hypertrophy and left ventricular dilatation. In addition, renal and cardiac infiltration of CD68 macrophages, intercellular adhesion molecule-1, and total collagen content were increased after Ang-II infusion in Tß4 KO mice (P<0.005). Overall, our data indicate that endogenous Tß4 is crucial in preventing tissue injury from Ang-II-induced hypertension. This study gives new insights into the protective role of endogenous Tß4 in hypertensive end-organ damage.


Subject(s)
Acute Kidney Injury/physiopathology , Blood Pressure/physiology , Cardiomyopathies/physiopathology , Hypertension/physiopathology , Thymosin/administration & dosage , Thymosin/deficiency , Acute Kidney Injury/metabolism , Acute Kidney Injury/prevention & control , Angiotensin II/toxicity , Animals , Cardiomyopathies/metabolism , Cardiomyopathies/prevention & control , Hypertension/chemically induced , Hypertension/metabolism , Infusions, Intravenous , Male , Mice , Mice, Knockout , Microfilament Proteins , Random Allocation , Rats
9.
PLoS One ; 13(1): e0190300, 2018.
Article in English | MEDLINE | ID: mdl-29364896

ABSTRACT

The natural peptide N-Acetyl-Seryl-Aspartyl-Lysyl-Proline (Ac-SDKP) decreases inflammation in chronic diseases such as hypertension and heart failure. However, Ac-SDKP effects on acute inflammatory responses during myocardial infarction (MI) are unknown. During the first 72 hours post-MI, neutrophils, M1 macrophages (pro-inflammatory), and M2 macrophages (pro-resolution) and release of myeloperoxidase (MPO) and matrix metalloproteinases (MMP) are involved in cardiac rupture. We hypothesized that in the acute stage of MI, Ac-SDKP decreases the incidence of cardiac rupture and mortality by preventing immune cell infiltration as well as by decreasing MPO and MMP expression. MI was induced by ligating the left descending coronary artery in C57BL/6 mice. Vehicle or Ac-SDKP (1.6 mg/kg/d) was infused via osmotic minipump. Cardiac immune cell infiltration was assessed by flow cytometry, cardiac MPO and MMP levels were measured at 24-48 hrs post-MI. Cardiac rupture and mortality incidence were determined at 7 days post-MI. In infarcted mice, Ac-SDKP significantly decreased cardiac rupture incidence from 51.0% (26 of 51 animals) to 27.3% (12 of 44) and mortality from 56.9% (29 of 51) to 31.8% (14 of 44). Ac-SDKP reduced M1 macrophages in cardiac tissue after MI, without affecting M2 macrophages and neutrophils. Ac-SDKP decreased MMP-9 activation in infarcted hearts with no changes on MPO expression. Ac-SDKP prevents cardiac rupture and decreases mortality post-acute MI. These protective effects of Ac-SDKP are associated with decreased pro-inflammatory M1 macrophage infiltration and MMP-9 activation.


Subject(s)
Heart Rupture/prevention & control , Myocardial Infarction/prevention & control , Oligopeptides/pharmacology , Animals , Chemotaxis, Leukocyte , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Macrophages/pathology , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Myocardial Infarction/mortality , Myocardial Infarction/pathology , Peroxidase/metabolism
10.
Am J Physiol Renal Physiol ; 315(4): F806-F811, 2018 10 01.
Article in English | MEDLINE | ID: mdl-28424211

ABSTRACT

Unilaterally nephrectomized rats (UNx) have higher glomerular capillary pressure (PGC) that can cause significant glomerular injury in the remnant kidney. PGC is controlled by the ratio of afferent (Af-Art) and efferent arteriole resistance. Af-Art resistance in turn is regulated by two intrinsic feedback mechanisms: 1) tubuloglomerular feedback (TGF) that causes Af-Art constriction in response to increased NaCl in the macula densa; and 2) connecting tubule glomerular feedback (CTGF) that causes Af-Art dilatation in response to an increase in NaCl transport in the connecting tubule via the epithelial sodium channel (ENaC). Resetting of TGF post-UNx can allow systemic pressure to be transmitted to the glomerulus and cause renal damage, but the mechanism behind this resetting is unclear. Since CTGF is an Af-Art dilatory mechanism, we hypothesized that CTGF is increased after UNx and contributes to TGF resetting. To test this hypothesis, we performed UNx in Sprague-Dawley (8) rats. Twenty-four hours after surgery, we performed micropuncture of individual nephrons and measured stop-flow pressure (PSF). PSF is an indirect measurement of PGC. Maximal TGF response at 40 nl/min was 8.9 ± 1.24 mmHg in sham-UNx rats and 1.39 ± 1.02 mmHg in UNx rats, indicating TGF resetting after UNx. When CTGF was inhibited with the ENaC blocker benzamil (1 µM/l), the TGF response was 12.29 ± 2.01 mmHg in UNx rats and 13.03 ± 1.25 mmHg in sham-UNx rats, indicating restoration of the TGF responses in UNx. We conclude that enhanced CTGF contributes to TGF resetting after UNx.


Subject(s)
Feedback , Kidney Glomerulus/blood supply , Kidney Tubules/blood supply , Nephrectomy , Nephrons/blood supply , Animals , Arterioles/physiology , Blood Pressure/physiology , Epithelial Sodium Channels/metabolism , Glomerular Filtration Rate/physiology , Kidney Tubules/physiology , Nephrectomy/methods , Rats, Sprague-Dawley
11.
Am J Physiol Renal Physiol ; 314(1): F99-F106, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28978533

ABSTRACT

Renal blood flow (RBF) provides important information regarding renal physiology and nephropathies. Arterial spin labeling-magnetic resonance imaging (ASL-MRI) is a noninvasive method of measuring blood flow without exogenous contrast media. However, low signal-to-noise ratio and respiratory motion artifacts are challenges for RBF measurements in small animals. Our objective was to evaluate the feasibility and reproducibility of RBF measurements by ASL-MRI using respiratory-gating and navigator correction methods to reduce motion artifacts. ASL-MRI images were obtained from the kidneys of Sprague-Dawley (SD) rats on a 7-Tesla Varian MRI system with a spin-echo imaging sequence. After 4 days, the study was repeated to evaluate its reproducibility. RBF was also measured in animals under unilateral nephrectomy and in renal artery stenosis (RST) to evaluate the sensitivity in high and low RBF models, respectively. RBF was also evaluated in Dahl salt-sensitive (SS) rats and spontaneous hypertensive rats (SHR). In SD rats, the cortical RBFs (cRBF) were 305 ± 59 and 271.8 ± 39 ml·min-1·100 g tissue-1 in the right and left kidneys, respectively. Retest analysis revealed no differences ( P = 0.2). The test-retest reliability coefficient was 92 ± 5%. The cRBFs before and after the nephrectomy were 296.8 ± 30 and 428.2 ± 45 ml·min-1·100 g tissue-1 ( P = 0.02), respectively. The kidneys with RST exhibited a cRBF decrease compared with sham animals (86 ± 17.6 vs. 198 ± 33.7 ml·min-1·100 g tissue-1; P < 0.01). The cRBFs in SD, Dahl-SS, and SHR rats were not different ( P = 0.35). We conclude that ASL-MRI performed with navigator correction and respiratory gating is a feasible and reliable noninvasive method for measuring RBF in rats.


Subject(s)
Image Processing, Computer-Assisted , Kidney Diseases/diagnostic imaging , Kidney Diseases/pathology , Magnetic Resonance Imaging , Animals , Contrast Media , Kidney/blood supply , Kidney/diagnostic imaging , Magnetic Resonance Imaging/methods , Male , Rats, Sprague-Dawley , Renal Artery/pathology , Renal Circulation/physiology , Spin Labels
12.
Hypertension ; 70(5): 982-989, 2017 11.
Article in English | MEDLINE | ID: mdl-28874464

ABSTRACT

The MAS1 receptor (R) exerts protective effects in the brain, heart, vessels, and kidney. R trafficking plays a critical function in signal termination and propagation and in R resensitization. We examined MAS1R internalization and trafficking on agonist stimulation and the role of ß-arrestin2 in the activation of ERK1/2 (extracellular signal-regulated kinase 1/2) and Akt after MAS1R stimulation. Human embryonic kidney 293T cells were transfected with the coding sequence for MAS1R-YFP (MAS1R fused to yellow fluorescent protein). MAS1R internalization was evaluated by measuring the MAS1R present in the plasma membrane after agonist stimulation using a ligand-binding assay. MAS1R trafficking was evaluated by its colocalization with trafficking markers. MAS1R internalization was blocked in the presence of shRNAcaveolin-1 and with dominant negatives for Eps15 (a protein involved in endocytosed Rs by clathrin-coated pits) and for dynamin. After stimulation, MAS1R colocalized with Rab11-a slow recycling vesicle marker-and not with Rab4-a fast recycling vesicle marker-or LysoTracker-a lysosome marker. Cells transfected with MAS1R showed an increase in Akt and ERK1/2 activation on angiotensin-(1-7) stimulation, which was blocked when the clathrin-coated pits pathway was blocked. Suppression of ß-arrestin2 by shRNA reduced the angiotensin-(1-7)-induced ERK1/2 activation, whereas Akt activation was not modified. We conclude that on agonist stimulation, MAS1R is internalized through clathrin-coated pits and caveolae in a dynamin-dependent manner and is then slowly recycled back to the plasma membrane. MAS1R induced Akt and ERK1/2 activation from early endosomes, and the activation of ERK1/2 was mediated by ß-arrestin2. Thus, MAS1R activity and density may be tightly controlled by the cell.


Subject(s)
Angiotensin I/metabolism , Endocytosis/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Peptide Fragments/metabolism , Protein Transport/physiology , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , beta-Arrestin 2/metabolism , Endosomes/physiology , HEK293 Cells , Humans , Proto-Oncogene Mas , Signal Transduction/physiology
13.
Am J Physiol Renal Physiol ; 313(6): F1209-F1215, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28835421

ABSTRACT

Afferent arteriole (Af-Art) resistance is modulated by two intrinsic nephron feedbacks: 1) the vasoconstrictor tubuloglomerular feedback (TGF) mediated by Na+-K+-2Cl- cotransporters (NKCC2) in the macula densa and blocked by furosemide and 2) the vasodilator connecting tubule glomerular feedback (CTGF), mediated by epithelial Na+ channels (ENaC) in the connecting tubule and blocked by benzamil. High salt intake reduces Af-Art vasoconstrictor ability in Dahl salt-sensitive rats (Dahl SS). Previously, we measured CTGF indirectly, by differences between TGF responses with and without CTGF inhibition. We recently developed a new method to measure CTGF more directly by simultaneously inhibiting NKCC2 and the Na+/H+ exchanger (NHE). We hypothesize that in vivo during simultaneous inhibition of NKCC2 and NHE, CTGF causes an Af-Art dilatation revealed by an increase in stop-flow pressure (PSF) in Dahl SS and that is enhanced with a high salt intake. In the presence of furosemide alone, increasing nephron perfusion did not change the PSF in either Dahl salt-resistant rats (Dahl SR) or Dahl SS. When furosemide and an NHE inhibitor, dimethylamiloride, were perfused simultaneously, an increase in tubular flow caused Af-Art dilatation that was demonstrated by an increase in PSF. This increase was greater in Dahl SS [4.5 ± 0.4 (SE) mmHg] than in Dahl SR (2.5 ± 0.3 mmHg; P < 0.01). We confirmed that CTGF causes this vasodilation, since benzamil completely blocked this effect. However, a high salt intake did not augment the Af-Art dilatation. We conclude that during simultaneous inhibition of NKCC2 and NHE in the nephron, CTGF induces Af-Art dilatation and a high salt intake failed to enhance this effect.


Subject(s)
Arterioles/physiopathology , Kidney Glomerulus/blood supply , Kidney Tubules/physiopathology , Renal Circulation , Sodium Chloride, Dietary/adverse effects , Vasodilation , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , Arterioles/drug effects , Arterioles/metabolism , Epithelial Sodium Channels/drug effects , Epithelial Sodium Channels/metabolism , Feedback, Physiological , Furosemide/pharmacology , Kidney Tubules/drug effects , Kidney Tubules/metabolism , Male , Rats, Inbred Dahl , Renal Circulation/drug effects , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Hydrogen Exchangers/metabolism , Solute Carrier Family 12, Member 1/antagonists & inhibitors , Solute Carrier Family 12, Member 1/metabolism , Time Factors , Vascular Resistance , Vasoconstriction , Vasodilation/drug effects
14.
Physiol Rep ; 5(2)2017 Jan.
Article in English | MEDLINE | ID: mdl-28126732

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune disease with a high prevalence of hypertension. NZBWF1 (SLE-Hyp) mice develop hypertension that can be prevented by modulating T cells. The peptide N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP) decreases renal damage and improves renal function in a model of SLE without hypertension (MRL/lpr). However, it is not known whether Ac-SDKP prevents hypertension in NZBWF1 mice. We hypothesized that in SLE-Hyp, Ac-SDKP prevents hypertension and renal damage by modulating T cells. Animals were divided into four groups: (1) control + vehicle, (2) control + Ac-SDKP, (3) SLE + vehicle, and (4) SLE + Ac-SDKP Systolic blood pressure (SBP), albuminuria, renal fibrosis, and T-cell phenotype were analyzed. SBP was higher in SLE compared to control mice and was not decreased by Ac-SDKP treatment. Half of SLE mice developed an acute and severe form of hypertension accompanied by albuminuria followed by death. Ac-SDKP delayed development of severe hypertension, albuminuria, and early mortality, but this delay did not reach statistical significance. Ac-SDKP prevented glomerulosclerosis, but not interstitial fibrosis in SLE-Hyp mice. SLE-Hyp mice showed a decrease in helper and cytotoxic T cells as well as an increase in double negative lymphocytes and T helper 17 cells, but these cells were unaffected by Ac-SDKP In conclusion, Ac-SDKP prevents kidney damage, without affecting blood pressure in an SLE animal model. However, during the acute relapse of SLE, Ac-SDKP might also delay the manifestation of an acute and severe form of hypertension leading to early mortality. Ac-SDKP is a potential tool to treat renal damage in SLE-Hyp mice.


Subject(s)
Hypertension/immunology , Kidney Diseases/immunology , Lupus Erythematosus, Systemic/physiopathology , Oligopeptides/administration & dosage , Albuminuria/prevention & control , Animals , Blood Pressure/drug effects , Female , Fibrosis/prevention & control , Hypertension/complications , Hypertension/prevention & control , Kidney/drug effects , Kidney/pathology , Kidney Diseases/complications , Kidney Diseases/pathology , Kidney Diseases/prevention & control , Lupus Erythematosus, Systemic/complications , Lupus Erythematosus, Systemic/mortality , Mice , Oligopeptides/therapeutic use , Survival Analysis , T-Lymphocytes/drug effects
15.
Hypertension ; 68(4): 1039-48, 2016 10.
Article in English | MEDLINE | ID: mdl-27550920

ABSTRACT

Bradykinin B2 receptor (B2R) and angiotensin-(1-7) Mas receptor (MasR)-mediated effects are physiologically interconnected. The molecular basis for such cross talk is unknown. It is hypothesized that the cross talk occurs at the receptor level. We investigated B2R-MasR heteromerization and the functional consequences of such interaction. B2R fused to the cyan fluorescent protein and MasR fused to the yellow fluorescent protein were transiently coexpressed in human embryonic kidney293T cells. Fluorescence resonance energy transfer analysis showed that B2R and MasR formed a constitutive heteromer, which was not modified by their agonists. B2R or MasR antagonists decreased fluorescence resonance energy transfer efficiency, suggesting that the antagonist promoted heteromer dissociation. B2R-MasR heteromerization induced an 8-fold increase in the MasR ligand-binding affinity. On agonist stimulation, the heteromer was internalized into early endosomes with a slower sequestration rate from the plasma membrane, compared with single receptors. B2R-MasR heteromerization induced a greater increase in arachidonic acid release and extracellular signal-regulated kinase phosphorylation after angiotensin-(1-7) stimulation, and this effect was blocked by the B2R antagonist. Concerning serine/threonine kinase Akt activity, a significant bradykinin-promoted activation was detected in B2R-MasR but not in B2R-expressing cells. Angiotensin-(1-7) and bradykinin elicited antiproliferative effects only in cells expressing B2R-MasR heteromers, but not in cells expressing each receptor alone. Proximity ligation assay confirmed B2R-MasR interaction in human glomerular endothelial cells supporting the interaction between both receptors in vivo. Our findings provide an explanation for the cross talk between bradykinin B2R and angiotensin-(1-7) MasR-mediated effects. B2R-MasR heteromerization induces functional changes in the receptor that may lead to long-lasting protective properties.


Subject(s)
Angiotensin I/metabolism , Bradykinin B2 Receptor Antagonists/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Peptide Fragments/metabolism , Receptor Cross-Talk/physiology , Receptor, Bradykinin B2/metabolism , Analysis of Variance , Angiotensin I/drug effects , Animals , Cell Membrane/metabolism , Cells, Cultured , HEK293 Cells , Humans , Peptide Fragments/drug effects , Proto-Oncogene Mas , Rats , Receptor Cross-Talk/drug effects , Receptor, Bradykinin B2/drug effects , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology , Sensitivity and Specificity , Transfection
16.
Am J Physiol Heart Circ Physiol ; 311(5): H1287-H1296, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27496875

ABSTRACT

Galectin-3 (Gal-3), a member of the ß-galactoside lectin family, has an important role in immune regulation. In hypertensive rats and heart failure patients, Gal-3 is considered a marker for an unfavorable prognosis. Nevertheless, the role and mechanism of Gal-3 action in hypertension-induced target organ damage are unknown. We hypothesized that, in angiotensin II (ANG II)-induced hypertension, genetic deletion of Gal-3 prevents left ventricular (LV) adverse remodeling and LV dysfunction by reducing the innate immune responses and myocardial fibrosis. To induce hypertension, male C57BL/6J and Gal-3 knockout (KO) mice were infused with ANG II (3 µg·min-1·kg-1 sc) for 8 wk. We assessed: 1) systolic blood pressure by plethysmography, 2) LV function and remodeling by echocardiography, 3) myocardial fibrosis by histology, 4) cardiac CD68+ macrophage infiltration by histology, 5) ICAM-1 and VCAM-1 expression by Western blotting, 6) plasma cytokines, including interleukin-6 (IL-6), by enzyme-linked immunosorbent assay, and 7) regulatory T (Treg) cells by flow cytometry as detected by their combined expression of CD4, CD25, and FOXP3. Systolic blood pressure and cardiac hypertrophy increased similarly in both mouse strains when infused with ANG II. However, hypertensive C57BL/6J mice suffered impaired ejection and shortening fractions. In these mice, the extent of myocardial fibrosis and macrophage infiltration was greater in histological sections, and cardiac ICAM-1, as well as plasma IL-6, expression was higher as assessed by Western blotting. However, all these parameters were blunted in Gal-3 KO mice. Hypertensive Gal-3 KO mice also had a higher number of splenic Treg lymphocytes. In conclusion, in ANG II-induced hypertension, genetic deletion of Gal-3 prevented LV dysfunction without affecting blood pressure or LV hypertrophy. This study indicates that the ANG II effects are, in part, mediated or triggered by Gal-3 together with the related intercellular signaling (ICAM-1 and IL-6), leading to cardiac inflammation and fibrosis.


Subject(s)
Angiotensin II/toxicity , Cardiomegaly/diagnostic imaging , Galectin 3/genetics , Hypertension/genetics , Macrophages/pathology , Myocardium/pathology , Ventricular Dysfunction, Left/diagnostic imaging , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Blood Pressure , Blotting, Western , Cardiomegaly/etiology , Cytokines/metabolism , Disease Models, Animal , Echocardiography , Enzyme-Linked Immunosorbent Assay , Fibrosis , Flow Cytometry , Hypertension/chemically induced , Hypertension/complications , Hypertension/physiopathology , Intercellular Adhesion Molecule-1/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/metabolism , Plethysmography , T-Lymphocytes, Regulatory , Vascular Cell Adhesion Molecule-1/metabolism , Ventricular Dysfunction, Left/etiology , Ventricular Function, Left
17.
Am J Physiol Renal Physiol ; 311(6): F1182-F1188, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27413197

ABSTRACT

Connecting tubule glomerular feedback (CTGF) is a mechanism where an increase in sodium (Na) concentration in the connecting tubule (CNT) causes the afferent arteriole (Af-Art) to dilate. We recently reported that aldosterone within the CNT lumen enhances CTGF via a nongenomic effect involving GPR30 receptors and sodium/hydrogen exchanger (NHE), but the signaling pathways of this mechanism are unknown. We hypothesize that aldosterone enhances CTGF via cAMP/protein kinase A (PKA) pathway that activates protein kinase C (PKC) and stimulates superoxide (O2-) production. Rabbit Af-Arts and their adherent CNTs were microdissected and simultaneously perfused. Two consecutive CTGF curves were elicited by increasing the CNT luminal NaCl. We found that the main effect of aldosterone was to sensitize CTGF and we analyzed data by comparing NaCl concentration in the CNT perfusate needed to achieve half of the maximal response (EC50). During the control period, the NaCl concentration that elicited a half-maximal response (EC50) was 37.0 ± 2.0 mmol/l; addition of aldosterone (10-8 mol/l) to the CNT lumen decreased EC50 to 19.3 ± 1.3 mmol/l (P ≤ 0.001 vs. Control). The specific adenylyl cyclase inhibitor 2',3'-dideoxyadenosine (ddA; 2 × 10-4 mol/l) and the PKA inhibitor H-89 dihydrochloride hydrate (H-89; 2 × 10-6 mol/l) prevented the aldosterone effect. The selective PKC inhibitor GF109203X (10-8 mol/l) also prevented EC50 reduction caused by aldosterone. CNT intraluminal addition of O2- scavenger tempol (10-4 mol/l) blocked the aldosterone effect. We conclude that aldosterone inside the CNT lumen enhances CTGF via a cAMP/PKA/PKC pathway and stimulates O2- generation and this process may contribute to renal damage by increasing glomerular capillary pressure.


Subject(s)
Aldosterone/pharmacology , Feedback, Physiological/drug effects , Kidney Glomerulus/drug effects , Signal Transduction/drug effects , Sodium Chloride/metabolism , Animals , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic N-Oxides/pharmacology , Dose-Response Relationship, Drug , Isoquinolines/pharmacology , Kidney Glomerulus/metabolism , Male , Protein Kinase C/metabolism , Rabbits , Spin Labels , Sulfonamides/pharmacology
18.
Am J Physiol Renal Physiol ; 310(10): F1026-34, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26962108

ABSTRACT

N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP) is a natural tetrapeptide with anti-inflammatory and antifibrotic properties. Previously, we have shown that prolyl oligopeptidase (POP) is involved in the Ac-SDKP release from thymosin-ß4 (Tß4). However, POP can only hydrolyze peptides shorter than 30 amino acids, and Tß4 is 43 amino acids long. This indicates that before POP hydrolysis takes place, Tß4 is hydrolyzed by another peptidase that releases NH2-terminal intermediate peptide(s) with fewer than 30 amino acids. Our peptidase database search pointed out meprin-α metalloprotease as a potential candidate. Therefore, we hypothesized that, prior to POP hydrolysis, Tß4 is hydrolyzed by meprin-α. In vitro, we found that the incubation of Tß4 with both meprin-α and POP released Ac-SDKP, whereas no Ac-SDKP was released when Tß4 was incubated with either meprin-α or POP alone. Incubation of Tß4 with rat kidney homogenates significantly released Ac-SDKP, which was blocked by the meprin-α inhibitor actinonin. In addition, kidneys from meprin-α knockout (KO) mice showed significantly lower basal Ac-SDKP amount, compared with wild-type mice. Kidney homogenates from meprin-α KO mice failed to release Ac-SDKP from Tß4. In vivo, we observed that rats treated with the ACE inhibitor captopril increased plasma concentrations of Ac-SDKP, which was inhibited by the coadministration of actinonin (vehicle, 3.1 ± 0.2 nmol/l; captopril, 15.1 ± 0.7 nmol/l; captopril + actinonin, 6.1 ± 0.3 nmol/l; P < 0.005). Similar results were obtained with urinary Ac-SDKP after actinonin treatment. We conclude that release of Ac-SDKP from Tß4 is mediated by successive hydrolysis involving meprin-α and POP.


Subject(s)
Kidney/metabolism , Metalloendopeptidases/metabolism , Oligopeptides/metabolism , Serine Endopeptidases/metabolism , Thymosin/metabolism , Animals , Blood Pressure , Captopril , Hydroxamic Acids , Male , Mice, Inbred C57BL , Mice, Knockout , Prolyl Oligopeptidases , Random Allocation , Rats, Sprague-Dawley
19.
Am J Physiol Heart Circ Physiol ; 310(9): H1176-83, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26945075

ABSTRACT

N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP) is a naturally occurring tetrapeptide that prevents inflammation and fibrosis in hypertension and other cardiovascular diseases. We previously showed that, in angiotensin II-induced hypertension, Ac-SDKP decreased the activation of nuclear transcription factor NF-κB, whereas, in experimental autoimmune myocarditis and hypertension animal models, it also reduced the expression of endothelial leukocyte adhesion molecule ICAM-1. However, the mechanisms by which Ac-SDKP downregulated ICAM-1 expression are still unclear. TNF-α is a proinflammatory cytokine that induces ICAM-1 expression in various cell types via TNF receptor 1 and activation of the classical NF-κB pathway. We hypothesized that in endothelial cells Ac-SDKP suppresses TNF-α-induced ICAM-1 expression by decreasing IKK phosphorylation that as a consequence leads to a decrease of IκB phosphorylation and NF-κB activation. To test this hypothesis, human coronary artery endothelial cells were treated with Ac-SDKP and then stimulated with TNF-α. We found that TNF-α-induced ICAM-1 expression was significantly decreased by Ac-SDKP in a dose-dependent manner. Ac-SDKP also decreased TNF-α-induced NF-κB translocation from cytosol to nucleus, as assessed by electrophoretic mobility shift assay, which correlated with a decrease in IκB phosphorylation. In addition, we found that Ac-SDKP decreased TNF-α-induced IKK phosphorylation and IKK-ß expression. However, Ac-SDKP had no effect on TNF-α-induced phosphorylation of p38 MAP kinase or ERK. Thus we conclude that Ac-SDKP inhibition of TNF-α activation of canonical, i.e., IKK-ß-dependent, NF-κB pathway and subsequent decrease in ICAM-1 expression is achieved via inhibition of IKK-ß.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Endothelial Cells/drug effects , I-kappa B Kinase/metabolism , Intercellular Adhesion Molecule-1/metabolism , NF-kappa B/metabolism , Oligopeptides/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Active Transport, Cell Nucleus/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Endothelial Cells/enzymology , Humans , Phosphorylation , Signal Transduction , Up-Regulation
20.
Am J Physiol Renal Physiol ; 310(8): F748-F754, 2016 04 15.
Article in English | MEDLINE | ID: mdl-26887831

ABSTRACT

In thick ascending limbs (THALs), nitric oxide (NO) decreases NaCl reabsorption via cGMP-mediated inhibition of Na-K-2Cl cotransporter (NKCC2). In angiotensin (ANG II)-induced hypertension, endothelin-1 (ET-1)-induced NO production by THALs is impaired. However, whether this alters NO's natriuretic effects and the mechanisms involved are unknown. In other cell types, ANG II augments phosphodiesterase 5 (PDE5)-mediated cGMP degradation. We hypothesized that NO-mediated inhibition of NKCC2 activity and stimulation of cGMP synthesis are blunted via PDE5 in ANG II-induced hypertension. Sprague-Dawley rats were infused with vehicle or ANG II (200 ng·kg-1·min-1) for 5 days. ET-1 reduced NKCC2 activity by 38 ± 13% (P < 0.05) in THALs from vehicle-treated rats but not from ANG II-hypertensive rats (Δ: -9 ± 13%). A NO donor yielded similar results as ET-1. In contrast, dibutyryl-cGMP significantly decreased NKCC2 activity in both vehicle-treated and ANG II-hypertensive rats (control: Δ-44 ± 15% vs. ANG II: Δ-41 ± 10%). NO increased cGMP by 2.08 ± 0.36 fmol/µg protein in THALs from vehicle-treated rats but only 1.06 ± 0.25 fmol/µg protein in ANG II-hypertensive rats (P < 0.04). Vardenafil (25 nM), a PDE5 inhibitor, restored NO's ability to inhibit NKCC2 activity in THALs from ANG II-hypertensive rats (Δ: -60 ± 9%, P < 0.003). Similarly, NO's stimulation of cGMP was also restored by vardenafil (vehicle-treated: 1.89 ± 0.71 vs. ANG II-hypertensive: 2.02 ± 0.32 fmol/µg protein). PDE5 expression did not differ between vehicle-treated and ANG II-hypertensive rats. We conclude that NO-induced inhibition of NKCC2 and increases in cGMP are blunted in ANG II-hypertensive rats due to PDE5 activation. Defects in the response of THALs to NO may enhance NaCl retention in ANG II-induced hypertension.


Subject(s)
Angiotensin II , Endothelin-1/pharmacology , Hypertension/metabolism , Loop of Henle/metabolism , Nitric Oxide/metabolism , Solute Carrier Family 12, Member 1/metabolism , Animals , Cyclic CMP/analogs & derivatives , Cyclic CMP/pharmacology , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Hypertension/chemically induced , Loop of Henle/drug effects , Male , Nitric Oxide Donors/pharmacology , Phosphodiesterase 5 Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Vardenafil Dihydrochloride/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...