Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Physiol Rep ; 7(24): e14302, 2019 12.
Article in English | MEDLINE | ID: mdl-31872559

ABSTRACT

Chymase released from mast cells produces pro-fibrotic, inflammatory, and vasoconstrictor agents. Studies were performed to test the hypothesis that chronic chymase inhibition provides a renal protective effect in type 2 diabetes. Diabetic (db/db) and control mice (db/m) were chronically infused with a chymase-specific inhibitor or vehicle for 8 weeks. Baseline urinary albumin excretion (UalbV) averaged 42 ± 3 and 442 ± 32 microg/d in control (n = 22) and diabetic mice (n = 27), respectively (p < .05). After administration of chymase inhibitor to diabetic mice, the change in UalbV was significantly lower (459 ± 57 microg/d) than in vehicle-treated diabetic mice (645 ± 108 microg/d). UNGAL V was not different at baseline between diabetic mice that would receive the chymase inhibitor (349 ± 56 ng/d, n = 6) and vehicle (373 ± 99 ng/d, n = 6) infusions, but increased significantly only in the vehicle-treated diabetic mice (p < .05). Glomeruli of diabetic kidneys treated chronically with chymase inhibition demonstrated reduced mesangial matrix expansion compared to glomeruli from untreated diabetic mice. Plasma angiotensin II levels were not altered by chymase inhibitor treatment. In summary, chronic chymase inhibition slowed the progression of urinary albumin excretion in diabetic mice. In conclusion, renal chymase may contribute to the progression of albuminuria in type 2 diabetes renal disease.


Subject(s)
Albuminuria/drug therapy , Chymases/antagonists & inhibitors , Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/drug therapy , Enzyme Inhibitors/therapeutic use , Oligopeptides/therapeutic use , Albuminuria/etiology , Animals , Chymases/metabolism , Diabetic Nephropathies/etiology , Enzyme Inhibitors/pharmacology , Kidney Glomerulus/drug effects , Kidney Glomerulus/metabolism , Male , Mice , Oligopeptides/pharmacology
2.
Am J Physiol Renal Physiol ; 311(2): F278-90, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27194718

ABSTRACT

In angiotensin II (ANG II)-dependent hypertension, there is an angiotensin type 1 receptor-dependent amplification mechanism enhancing intrarenal angiotensinogen (AGT) formation and secretion in the tubular fluid. To evaluate the role of increased arterial pressure, AGT mRNA, protein expression, and urinary AGT (uAGT) excretion and tissue injury were assessed in both kidneys of two-kidney, one-clip Sprague-Dawley hypertensive rats subjected to left renal arterial clipping (0.25-mm gap). By 18-21 days, systolic arterial pressure increased to 180 ± 3 mmHg, and uAGT increased. Water intake, body weights, 24-h urine volumes, and sodium excretion were similar. In separate measurements of renal function in anesthetized rats, renal plasma flow and glomerular filtration rate were similar in clipped and nonclipped kidneys and not different from those in sham rats, indicating that the perfusion pressure to the clipped kidneys remained within the autoregulatory range. The nonclipped kidneys exhibited increased urine flow and sodium excretion. The uAGT excretion was significantly greater in nonclipped kidneys compared with clipped and sham kidneys. AGT mRNA was 2.15-fold greater in the nonclipped kidneys compared with sham (1.0 ± 0.1) or clipped (0.98 ± 0.15) kidneys. AGT protein levels were also greater in the nonclipped kidneys. The nonclipped kidneys exhibited greater glomerular expansion and immune cell infiltration, medullary fibrosis, and cellular proliferation than the clipped kidneys. Because both kidneys have elevated ANG II levels, the greater tissue injury in the nonclipped kidneys indicates that an increased arterial pressure synergizes with increased intrarenal ANG II to stimulate AGT production and exert greater renal injury.


Subject(s)
Angiotensinogen/biosynthesis , Angiotensinogen/urine , Hypertension, Renovascular/pathology , Hypertension, Renovascular/urine , Kidney/metabolism , Kidney/pathology , Animals , Arterial Pressure , Body Weight , Drinking , Fibrosis , Immunity, Cellular , Kidney Glomerulus/pathology , Kidney Medulla/pathology , Male , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Sodium/urine
3.
Physiol Rep ; 3(10)2015 Oct.
Article in English | MEDLINE | ID: mdl-26486161

ABSTRACT

Angiotensin II (AngII) is a critical physiologic regulator of volume homeostasis and mean arterial pressure (MAP), yet it also is known to induce immune mechanisms that contribute to hypertension. This study determined the role of interleukin-6 (IL-6) in the physiologic effect of AngII to maintain normal MAP during low-salt (LS) intake, and whether hypertension induced by plasma AngII concentrations measured during LS diet required IL-6. IL-6 knockout (KO) and wild-type (WT) mice were placed on LS diet for 7 days, and MAP was measured 19 h/day with telemetry. MAP was not affected by LS in either group, averaging 101 ± 4 and 100 ± 4 mmHg in WT and KO mice, respectively, over the last 3 days. Seven days of ACEI decreased MAP ~25 mmHg in both groups. In other KO and WT mice, AngII was infused at 200 ng/kg per minute to approximate plasma AngII levels during LS. Surgical reduction of kidney mass and high-salt diet were used to amplify the blood pressure effect. The increase in MAP after 7 days was not different, averaging 20 ± 5 and 22 ± 6 mmHg in WT and KO mice, respectively. Janus Kinase 2 (JAK2)/signal transducer of activated transcription (STAT3) phosphorylation were not affected by LS, but were increased by AngII infusion at 200 and 800 ng/kg per minute. These data suggest that physiologic levels of AngII do not activate or require IL-6 to affect blood pressure significantly, whether AngII is maintaining blood pressure on LS diet or causing blood pressure to increase. JAK2/STAT3 activation, however, is tightly associated with AngII hypertension, even when caused by physiologic levels of AngII.

4.
Am J Physiol Renal Physiol ; 309(10): F880-8, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26268270

ABSTRACT

In contrast to the negative feedback of angiotensin II (ANG II) on juxtaglomerular renin, ANG II stimulates renin in the principal cells of the collecting duct (CD) in rats and mice via ANG II type 1 (AT1R) receptor, independently of blood pressure. In vitro data indicate that CD renin is augmented by AT1R activation through protein kinase C (PKC), but the exact mechanisms are unknown. We hypothesize that ANG II stimulates CD renin synthesis through AT1R via PKC and the subsequent activation of cAMP/PKA/CREB pathway. In M-1 cells, ANG II increased cAMP, renin mRNA (3.5-fold), prorenin, and renin proteins, as well as renin activity in culture media (2-fold). These effects were prevented by PKC inhibition with calphostin C, PKC-α dominant negative, and by PKA inhibition. Forskolin-induced increases in cAMP and renin expression were prevented by calphostin C. PKC inhibition and Ca2+ depletion impaired ANG II-mediated CREB phosphorylation and upregulation of renin. Adenylate cyclase 6 (AC) siRNA remarkably attenuated the ANG II-dependent upregulation of renin mRNA. Physiological activation of AC with vasopressin increased renin expression in M-1 cells. The results suggest that the ANG II-dependent upregulation of renin in the CD depends on PKC-α, which allows the augmentation of cAMP production and activation of PKA/CREB pathway via AC6. This study defines the intracellular signaling pathway involved in the ANG II-mediated stimulation of renin in the CD. This is a novel mechanism responsible for the regulation of local renin-angiotensin system in the distal nephron.


Subject(s)
Angiotensin II/pharmacology , Cyclic AMP Response Element-Binding Protein/metabolism , Protein Kinase C-alpha/metabolism , Renin/metabolism , Animals , Blood Pressure/drug effects , Mice , Phosphorylation , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology , Signal Transduction/physiology , Up-Regulation/drug effects
5.
Hypertension ; 65(4): 813-20, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25624342

ABSTRACT

Transcription factor E26 transformation-specific sequence-1 (ETS-1) is a transcription factor that regulates the expression of a variety of genes, including growth factors, chemokines, and adhesion molecules. We recently demonstrated that angiotensin II increases the glomerular expression of ETS-1 and that blockade of ETS-1 ameliorates the profibrotic and proinflammatory effects of angiotensin II. The Dahl salt-sensitive rat is a paradigm of salt-sensitive hypertension associated with local activation of the renin-angiotensin system. In these studies, we determined whether: (1) salt-sensitive hypertension is associated with renal expression of ETS-1 and (2) ETS-1 participates in the development of end-organ injury in salt-sensitive hypertension. Dahl salt-sensitive rats were fed a normal-salt diet (0.5% NaCl diet) or a high-salt diet (4% NaCl) for 4 weeks. Separate groups on high-salt diet received an ETS-1 dominant-negative peptide (10 mg/kg/d), an inactive ETS-1 mutant peptide (10 mg/kg/d), the angiotensin II type 1 receptor blocker candesartan (10 mg/kg/d), or the combination high-salt diet/dominant-negative peptide/angiotensin II type 1 receptor blocker for 4 weeks. High-salt diet rats had a significant increase in the glomerular expression of the phosphorylated ETS-1 that was prevented by angiotensin II type 1 receptor blocker. ETS-1 blockade reduced proteinuria, glomerular injury score, fibronectin expression, urinary transforming growth factor-ß excretion, and macrophage infiltration. Angiotensin II type 1 receptor blocker reduced proteinuria, glomerular injury score, and macrophage infiltration, whereas concomitant ETS-1 blockade and angiotensin II type 1 receptor blocker had additive effects and reduced interstitial fibrosis. Our studies demonstrated that salt-sensitive hypertension results in increased glomerular expression of phosphorylated ETS-1 and suggested that ETS-1 plays an important role in the pathogenesis of end-organ injury in salt-sensitive hypertension.


Subject(s)
Acute Kidney Injury/genetics , Angiotensins/metabolism , DNA/genetics , Gene Expression Regulation , Hypertension/genetics , Kidney Glomerulus/metabolism , Proto-Oncogene Protein c-ets-1/genetics , Acute Kidney Injury/etiology , Acute Kidney Injury/metabolism , Animals , Blotting, Western , Disease Models, Animal , Hypertension/complications , Hypertension/metabolism , Immunohistochemistry , Kidney Glomerulus/pathology , Male , Proto-Oncogene Protein c-ets-1/biosynthesis , Rats , Rats, Inbred Dahl
6.
Am J Med Sci ; 348(5): 416-22, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25250989

ABSTRACT

BACKGROUND: Evidence indicates that chronic angiotensin II (AngII) infusion increases (pro)renin receptor ((P)RR) expression in renal inner medullary collecting duct (IMCD) cells. Recently, it has been shown that renal (P)RR expression is augmented during a low-salt (LS) diet. However, the role of AngII in mediating the stimulation of (P)RR during LS conditions is unknown. We hypothesized that AngII mediates the increased expression of (P)RR during low-salt conditions in IMCDs. METHODS: (P)RR expression and AngII levels were evaluated in Sprague-Dawley rats fed a LS diet (0.03% NaCl) and normal salt (NS; 0.4% NaCl) for 7 days. We examined the effects of sodium reduction (130 mM NaCl) and AngII on (P)RR expression in IMCDs isolated in hypertonic conditions (640 mOsmol/L with 280 mM NaCl). RESULTS: Plasma renin activity in LS rats was significantly higher than rats fed with NS (28.1 ± 2.2 versus 6.7 ± 1.1 ng AngI·mL⁻¹·hr⁻¹; P < 0.05), as well as renin content in renal cortex and medulla. The (P)RR mRNA and protein levels were higher in medullary tissues from LS rats but did not change in the cortex. Intrarenal AngII was augmented in LS compared with NS rats (cortex: 710 ± 113 versus 277 ± 86 fmol/g, P < 0.05; medulla: 2093 ± 125 versus 1426 ± 126 fmol/g, P < 0.05). In cultured IMCDs, (P)RR expression was increased in response to LS or AngII treatment and potentiated by both treatments (both at 640 mOsmol/L). CONCLUSIONS: These data indicate that (P)RR is augmented in medullary collecting ducts in response to LS and that this effect is further enhanced by the increased intrarenal AngII content.


Subject(s)
Angiotensin II/pharmacology , Diet, Sodium-Restricted , Gene Expression Regulation , Receptors, Cell Surface/biosynthesis , Animals , Diet, Sodium-Restricted/methods , Kidney Tubules, Collecting/metabolism , Male , Rats , Rats, Sprague-Dawley , Prorenin Receptor
7.
J Am Soc Nephrol ; 25(12): 2752-63, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25012170

ABSTRACT

The kidney is an important source of angiotensin-converting enzyme (ACE) in many species, including humans. However, the specific effects of local ACE on renal function and, by extension, BP control are not completely understood. We previously showed that mice lacking renal ACE, are resistant to the hypertension induced by angiotensin II infusion. Here, we examined the responses of these mice to the low-systemic angiotensin II hypertensive model of nitric oxide synthesis inhibition with L-NAME. In contrast to wild-type mice, mice without renal ACE did not develop hypertension, had lower renal angiotensin II levels, and enhanced natriuresis in response to L-NAME. During L-NAME treatment, the absence of renal ACE was associated with blunted GFR responses; greater reductions in abundance of proximal tubule Na(+)/H(+) exchanger 3, Na(+)/Pi co-transporter 2, phosphorylated Na(+)/K(+)/Cl(-) cotransporter, and phosphorylated Na(+)/Cl(-) cotransporter; and greater reductions in abundance and processing of the γ isoform of the epithelial Na(+) channel. In summary, the presence of ACE in renal tissue facilitates angiotensin II accumulation, GFR reductions, and changes in the expression levels and post-translational modification of sodium transporters that are obligatory for sodium retention and hypertension in response to nitric oxide synthesis inhibition.


Subject(s)
Hypertension/metabolism , Kidney/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Peptidyl-Dipeptidase A/physiology , Angiotensin II/metabolism , Animals , Blood Pressure , Glomerular Filtration Rate , Hypertension/drug therapy , Male , Mice , Mice, Inbred C57BL , NG-Nitroarginine Methyl Ester/chemistry , Natriuresis , Nitric Oxide/metabolism , Phosphorylation , Promoter Regions, Genetic , Protein Processing, Post-Translational , Renin/blood , Symporters/metabolism
8.
J Clin Invest ; 123(5): 2011-23, 2013 May.
Article in English | MEDLINE | ID: mdl-23619363

ABSTRACT

Activation of the intrarenal renin-angiotensin system (RAS) can elicit hypertension independently from the systemic RAS. However, the precise mechanisms by which intrarenal Ang II increases blood pressure have never been identified. To this end, we studied the responses of mice specifically lacking kidney angiotensin-converting enzyme (ACE) to experimental hypertension. Here, we show that the absence of kidney ACE substantially blunts the hypertension induced by Ang II infusion (a model of high serum Ang II) or by nitric oxide synthesis inhibition (a model of low serum Ang II). Moreover, the renal responses to high serum Ang II observed in wild-type mice, including intrarenal Ang II accumulation, sodium and water retention, and activation of ion transporters in the loop of Henle (NKCC2) and distal nephron (NCC, ENaC, and pendrin) as well as the transporter activating kinases SPAK and OSR1, were effectively prevented in mice that lack kidney ACE. These findings demonstrate that ACE metabolism plays a fundamental role in the responses of the kidney to hypertensive stimuli. In particular, renal ACE activity is required to increase local Ang II, to stimulate sodium transport in loop of Henle and the distal nephron, and to induce hypertension.


Subject(s)
Hypertension/metabolism , Kidney/metabolism , Peptidyl-Dipeptidase A/metabolism , Angiotensin II/metabolism , Animals , Kidney/embryology , Liver/metabolism , Loop of Henle/metabolism , Male , Mice , NG-Nitroarginine Methyl Ester/pharmacology , Protein Serine-Threonine Kinases/metabolism , Receptors, Drug/metabolism , Renin-Angiotensin System , Sodium/metabolism , Sodium-Potassium-Chloride Symporters/metabolism , Solute Carrier Family 12, Member 1 , Solute Carrier Family 12, Member 3 , Symporters/metabolism , Water/metabolism
9.
PLoS One ; 8(3): e58339, 2013.
Article in English | MEDLINE | ID: mdl-23516464

ABSTRACT

The binding of renin or prorenin to the (pro)renin receptor (PRR) promotes angiotensin (Ang) II formation and mediates Ang II-independent signaling pathways. In the central nervous system (CNS), Ang II regulates blood pressure via inducing oxidative stress; however, the role of PRR-mediated Ang II-independent signaling pathways in oxidative stress in the CNS remains undefined. To address this question, Neuro-2A cells were infected with control virus or an adeno-associated virus encoding the human PRR. Human PRR over-expression alone increased ROS levels, NADPH oxidase activity, as well as NADPH oxidase (NOX) isoforms 2 and 4 mRNA expression levels and these effects were not blocked by losartan. Moreover, the increase in NOX 2 and NOX 4 mRNA levels, NADPH oxidase activity, and ROS levels induced by PRR over-expression was prevented by mitogen activated protein kinase/extracellular signal-regulated kinase 1 and 2 (MAPK/ERK1/2) inhibition, and phosphoinositide 3 kinase/Akt (IP3/Akt) inhibition, indicating that PRR regulates NOX activity and ROS formation in neuro-2A cells through Ang II-independent ERK1/2 and IP3/Akt activation. Interestingly, at a concentration of 2 nM or higher, prorenin promoted Ang II formation, and thus further increased the ROS levels in cultured Neuro-2A cells via PRR. In conclusion, human PRR over-expression induced ROS production through both angiotensin II-dependent and -independent mechanisms. We showed that PRR-mediated angiotensin II-independent ROS formation is associated with activation of the MAPK/ERK1/2 and PI3/Akt signaling pathways and up-regulation of mRNA level of NOX 2 and NOX4 isoforms in neuronal cells.


Subject(s)
Angiotensin II/metabolism , Neurons/metabolism , Oxidative Stress , Receptors, Cell Surface/metabolism , Animals , Cell Line , Dependovirus/genetics , Gene Expression , Genetic Vectors/genetics , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , NADPH Oxidase 2 , NADPH Oxidase 4 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Receptors, Cell Surface/genetics , Renin/metabolism , Signal Transduction , Prorenin Receptor
10.
Am J Physiol Renal Physiol ; 304(5): F505-14, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23303412

ABSTRACT

In angiotensin II (ANG II) infusion hypertension, there is an augmentation of intratubular angiotensinogen (AGT) and ANG II leading to increased urinary AGT and ANG II excretion rates associated with tissue injury. However, the changes in urinary AGT and ANG II excretion rates and markers of renal injury during physiologically induced stimulation of the renin-angiotensin system (RAS) by a low-salt diet remain unclear. Male Sprague-Dawley rats received a low-salt diet (0.03% NaCl; n = 6) and normal-salt diet (0.3% NaCl, n = 6) for 13 days. Low-salt diet rats had markedly higher plasma renin activity and plasma ANG II levels. Kidney cortex renin mRNA, kidney AGT mRNA, and AGT immunoreactivity were not different; however, medullary renin mRNA, kidney renin content, and kidney ANG II levels were significantly elevated by the low-salt diet. Kidney renin immunoreactivity was also markedly increased in juxtaglomerular apparati and in cortical and medullary collecting ducts. Urinary AGT excretion rates and urinary ANG II excretion rates were not augmented by the low-salt diet. The low-salt diet caused mild renal fibrosis in glomeruli and the tubulointerstitium, but no other signs of kidney injury were evident. These results indicate that, in contrast to the response in ANG II infusion hypertension, the elevated plasma and intrarenal ANG II levels caused by physiological stimulation of RAS are not reflected by increased urinary AGT or ANG II excretion rates or the development of renal injury.


Subject(s)
Angiotensin II/metabolism , Angiotensinogen/metabolism , Kidney Tubules/metabolism , Renin-Angiotensin System/physiology , Sodium Chloride, Dietary/metabolism , Angiotensinogen/genetics , Animals , Blood Pressure/physiology , Male , Rats , Rats, Sprague-Dawley , Renin/genetics , Renin/metabolism
11.
Gend Med ; 9(4): 207-18, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22795463

ABSTRACT

BACKGROUND: The intrarenal renin-angiotensin system contributes to hypertension by regulating sodium and water reabsorption throughout the nephron. Sex differences in the intrarenal components of the renin-angiotensin system have been involved in the greater incidence of high blood pressure and progression to kidney damage in males than females. OBJECTIVE: This study investigated whether there is a sex difference in the intrarenal gene expression and urinary excretion of angiotensinogen (AGT) during angiotensin II (Ang II)-dependent hypertension and high-salt (HS) diet. METHODS: Male and female Sprague-Dawley rats were divided into 5 groups for each sex: Normal-salt control, HS diet (8% NaCl), Ang II-infused (80 ng/min), Ang II-infused plus HS diet, and Ang II-infused plus HS diet and treatment with the Ang II receptor blocker, candesartan (25 mg/L in the drinking water). Rats were evaluated for systolic blood pressure (SBP), kidney AGT mRNA expression, urinary AGT excretion, and proteinuria at different time points during a 14-day protocol. RESULTS: Both male and female rats exhibited similar increases in urinary AGT, with increases in SBP during chronic Ang II infusion. HS diet greatly exacerbated the urinary AGT excretion in Ang II-infused rats; males had a 9-fold increase over Ang II alone and females had a 2.5-fold increase. Male rats displayed salt-sensitive SBP increases during Ang II infusion and HS diet, and female rats did not. In the kidney cortex, males displayed greater AGT gene expression than females during all treatments. During Ang II infusion, both sexes exhibited increases in AGT gene message compared with same-sex controls. In addition, HS diet combined with Ang II infusion exacerbated the proteinuria in both sexes. Concomitant Ang II receptor blocker treatment during Ang II infusion and HS diet decreased SBP and urinary AGT similarly in both sexes; however, the decrease in proteinuria was greater in the females. CONCLUSION: During Ang II-dependent hypertension and HS diet, higher intrarenal renin-angiotensin system activation in males, as reflected by higher AGT gene expression and urinary excretion, indicates a mechanism for greater progression of high blood pressure and might explain the sex disparity in development of salt-sensitive hypertension.


Subject(s)
Angiotensin II/metabolism , Angiotensinogen/urine , Hypertension/physiopathology , Kidney/physiopathology , Sex Characteristics , Sodium Chloride, Dietary , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Hemodynamics/drug effects , Male , Oxidative Stress , Rats , Rats, Sprague-Dawley , Sex Factors
12.
Int J Hypertens ; 2012: 290635, 2012.
Article in English | MEDLINE | ID: mdl-23316344

ABSTRACT

It is well known that the brain renin-angiotensin (RAS) system plays an essential role in the development of hypertension, mainly through the modulation of autonomic activities and vasopressin release. However, how the brain synthesizes angiotensin (Ang) II has been a debate for decades, largely due to the low renin activity. This paper first describes the expression of the vasoconstrictive arm of RAS components in the brain as well as their physiological and pathophysiological significance. It then focus on the (pro)renin receptor (PRR), a newly discovered component of the RAS which has a high level in the brain. We review the role of prorenin and PRR in peripheral organs and emphasize the involvement of brain PRR in the pathogenesis of hypertension. Some future perspectives in PRR research are heighted with respect to novel therapeutic target for the treatment of hypertension and other cardiovascular diseases.

13.
Am J Physiol Renal Physiol ; 301(6): F1195-201, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21865264

ABSTRACT

Renin expression in principal cells of collecting ducts (CD) is upregulated in angiotensin II (ANG II)-dependent hypertensive rats; however, it remains unclear whether increased CD-derived renin undergoes tubular secretion. Accordingly, urinary levels of renin (uRen), angiotensinogen (uAGT), and ANG II (uANG II) were measured in chronic ANG II-infused Sprague-Dawley rats (80 ng/min for 14 days, n = 10) and sham-operated rats (n = 10). Systolic blood pressure increased in the ANG II rats by day 5 and continued to increase throughout the study (day 13; ANG II: 175 ± 10 vs. sham: 116 ± 2 mmHg; P < 0.05). ANG II infusion increased renal cortical and medullary ANG II levels (cortical ANG II: 606 ± 72 vs. 247 ± 43 fmol/g; P < 0.05; medullary ANG II: 2,066 ± 116 vs. 646 ± 36 fmol/g; P < 0.05). Although plasma renin activity (PRA) was suppressed in the ANG II-infused rats (0.3 ± 0.2 vs. 5.5 ± 1.8 ng ANG I·ml(-1)·h(-1); P < 0.05), renin content in renal medulla was increased (12,605 ± 1,343 vs. 7,956 ± 765 ng ANG I·h(-1)·mg(-1); P < 0.05). Excretion of uAGT and uANG II increased in the ANG II rats [uAGT: 1,107 ± 106 vs. 60 ± 26 ng/day; P < 0.0001; uANG II: 3,813 ± 431 vs. 2,080 ± 361 fmol/day; P < 0.05]. By day 13, despite suppression of PRA, urinary prorenin content increased in ANG II rats [15.7 ± 3 vs. 2.6 ± 1 × 10(-3) enzyme units excreted (EUE)/day, P < 0.01] as was the excretion rate of renin (8.6 ± 2 × 10(-6) EUE/day) compared with sham (2.8 ± 1 × 10(-6) EUE/day; P < 0.05). Urinary renin and prorenin protein levels examined by Western blot were augmented ∼10-fold in the ANG II-infused rats. Concomitant AT(1) receptor blockade with candesartan prevented the increase. Thus, in ANG II-dependent hypertensive rats with marked PRA suppression, increased urinary levels of renin and prorenin reflect their augmented secretion by CD cells into the luminal fluid. The greater availability of renin and AGT in the urine reflects the capability for intratubular ANG II formation which stimulates sodium reabsorption in distal nephron segments.


Subject(s)
Angiotensin II/urine , Angiotensinogen/urine , Renin/urine , Angiotensin II/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Benzimidazoles/pharmacology , Biphenyl Compounds , Blood Pressure/drug effects , Kidney/chemistry , Male , Rats , Rats, Sprague-Dawley , Renin/analysis , Renin/blood , Tetrazoles/pharmacology
14.
Hypertension ; 57(3): 594-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21282553

ABSTRACT

Collecting duct (CD) renin is stimulated by angiotensin (Ang) II, providing a pathway for Ang I generation and further conversion to Ang II. Ang II stimulates the epithelial sodium channel via the Ang II type 1 receptor and increases mineralocorticoid receptor activity attributed to increased aldosterone release. Our objective was to determine whether CD renin augmentation is mediated directly by Ang II type 1 receptor or via the epithelial sodium channel and mineralocorticoid receptor. In vivo studies examined the effects of epithelial sodium channel blockade (amiloride; 5 mg/kg per day) on CD renin expression and urinary renin content in Ang II-infused rats (80 ng/min, 2 weeks). Ang II infusion increased systolic blood pressure, medullary renin mRNA, urinary renin content, and intrarenal Ang II levels. Amiloride cotreatment did not alter these responses despite a reduction in the rate of progression of systolic blood pressure. In primary cultures of inner medullary CD cells, renin mRNA and (pro)renin protein levels increased with Ang II (100 nmol/L), and candesartan (Ang II type 1 receptor antagonist) prevented this effect. Aldosterone (10(-10) to 10(-7) mol/L) with or without amiloride did not modify the upregulation of renin mRNA in Ang II-treated cells. However, inhibition of protein kinase C with calphostin C prevented the Ang II-mediated increases in renin mRNA and (pro)renin protein levels. Furthermore, protein kinase C activation with phorbol 12-myristate 13-acetate increased renin expression to the same extent as Ang II. These data indicate that an Ang II type 1 receptor-mediated increase in CD renin is induced directly by Ang II via the protein kinase C pathway and that this regulation is independent of mineralocorticoid receptor activation or epithelial sodium channel activity.


Subject(s)
Angiotensin II/pharmacology , Epithelial Sodium Channels/metabolism , Kidney Medulla/metabolism , Protein Kinase C/metabolism , Receptors, Mineralocorticoid/metabolism , Renin/metabolism , Signal Transduction/physiology , Analysis of Variance , Angiotensin II/metabolism , Animals , Blotting, Western , Cells, Cultured , Fluorescent Antibody Technique , Hypertension/chemically induced , Hypertension/metabolism , Kidney Medulla/cytology , Kidney Medulla/drug effects , Male , Rats , Rats, Sprague-Dawley , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects
15.
Hypertension ; 57(4): 859-64, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21321306

ABSTRACT

Renin synthesis and secretion by principal cells of the collecting duct are enhanced in angiotensin (Ang) II-dependent hypertension. The presence of renin/(pro)renin and its receptor, the (pro)renin receptor ([P]RR), in the collecting duct may provide a pathway for Ang I generation with further conversion to Ang II. To assess whether (P)RR activation occurs during Ang II-dependent hypertension, we examined renal (P)RR levels and soluble (P)RR excretion in the urine of chronic Ang II-infused rats (80 ng/min; for 2 weeks; n=10) and sham-operated rats (n=10). Systolic blood pressure and Ang II levels in the plasma and kidney were increased whereas plasma renin activity was suppressed in Ang II-infused rats. Renal (P)RR transcripts were upregulated in the cortex and medulla of Ang II-infused rats. (P)RR immunoreactivity in collecting duct cells and the protein levels of the full-length form (37-kDa band) were significantly decreased in the medulla of Ang II-infused rats. The soluble (P)RR (28-kDa band) was detected in the renal medulla and urine samples of Ang II-infused rats, which also showed increases in urinary renin content. To determine whether the soluble (P)RR could stimulate Ang I formation, urine samples were incubated with recombinant human (pro)renin. Urine samples of Ang II-infused rats exhibited increased Ang I formation compared with sham-operated rats. Thus, in chronic Ang II-infused rats, the catalytic activity of the augmented renin produced in the collecting duct may be enhanced by the intraluminal soluble (P)RR and cell-surface located (P)RR, thus contributing to enhanced intratubular Ang II formation.


Subject(s)
Angiotensin II/pharmacology , Hypertension/metabolism , Kidney/metabolism , Receptors, Cell Surface/metabolism , Renin/blood , Analysis of Variance , Animals , Blood Pressure/physiology , Blotting, Western , Body Weight/physiology , Hypertension/chemically induced , Immunohistochemistry , Infusion Pumps, Implantable , Male , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Prorenin Receptor
16.
Circ Res ; 107(7): 934-8, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20689062

ABSTRACT

RATIONALE: Despite overwhelming evidence of the importance of brain renin-angiotensin system (RAS), the very existence of intrinsic brain RAS remains controversial. OBJECTIVE: To investigate the hypothesis that the brain (pro)renin receptor (PRR) is physiologically important in the brain RAS regulation and cardiovascular functions. METHODS AND RESULTS: PRR is broadly distributed within neurons of cardiovascular-relevant brain regions. The physiological functions of PRR were studied in the supraoptic nucleus (SON) because this brain region showed greater levels of PRR mRNA in the spontaneously hypertensive rats (SHR) compared with normotensive Wistar-Kyoto (WKY) rats. Adeno-associated virus (AAV)-mediated overexpression of human PRR in the SON of normal rats resulted in increases in plasma and urine vasopressin, and decreases in H(2)O intake and urine output without any effects on mean arterial pressure and heart rate. Knockdown of endogenous PRR by AAV-short hairpin RNA in the SON of SHRs attenuated age-dependent increases in mean arterial pressure and caused a decrease in heart rate and plasma vasopressin. Incubation of neuronal cells in culture with human prorenin and angiotensinogen resulted in increased generation of angiotensin I and II. Furthermore, renin treatment increased phosphorylation of extracellular signal-regulated kinase ½ in neurons from both WKY rats and SHRs; however, the stimulation was 50% greater in the SHR. CONCLUSIONS: The study demonstrates that brain PRR is functional and plays a role in the neural control of cardiovascular functions. This may help resolve a long-held controversy concerning the existence of intrinsic and functional brain RAS.


Subject(s)
Cardiovascular System/innervation , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Renin-Angiotensin System/physiology , Supraoptic Nucleus/physiology , Animals , Blood Pressure/physiology , Gene Knockdown Techniques , Green Fluorescent Proteins/genetics , Homeostasis/physiology , Hypertension/physiopathology , Male , RNA, Messenger/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Rats, Sprague-Dawley , Prorenin Receptor
17.
Hypertension ; 56(3): 378-83, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20625079

ABSTRACT

Rats infused chronically with Val(5)-Angiotensin (Ang) II exhibit increased urinary excretion of endogenous Ile(5)-Ang II by the 12th day of infusion, suggesting the stimulation of endogenous Ang II formation by Val(5)-Ang II infusion. The present study determined the time course of increased urinary Ang II excretion and the effects of Ang II type 1 receptor blockade (candesartan, 2 mg/kg per day) on the urinary excretion rates of Ile(5)-Ang II in Val(5)-Ang II-infused (80 ng/min) rats. Ile(5)-Ang II was separated from Val(5)-Ang II by high-performance liquid chromatography and measured by radioimmunoassay. Systolic blood pressure increased progressively (215+/-2 mm Hg) in Val(5)-Ang II-infused rats (n=5), whereas the candesartan-treated group (n=6) remained normotensive (124+/-3 mm Hg). Candesartan treatment significantly increased the level of plasma Ile(5)-Ang II (24.0+/-7.6 versus 156.9+/-24.6 fmol/mL; P<0.01); in contrast, there was a markedly lower intrarenal Ile(5)-Ang II content (357.9+/-76.6 versus 21.1+/-2.8 fmol/g; P<0.01). Urinary Ile(5)-Ang II excretion rates were elevated by day 9 (2185.7+/-283.2 fmol/24 hours) in Val(5)-Ang II-infused rats but not in candesartan-treated rats (740.6+/-110.3 fmol/24 hours). Thus, Ang II type 1 receptor blockade prevents the increase in urinary excretion of endogenous Ang II in rats subjected to chronic Ang II infusion. These data indicate that the increased urinary excretion of endogenous Ang II in Val(5)-Ang II-infused rats is primarily attributed to Ang II type 1 receptor-dependent secretion into and/or de novo formation of Ang II within the tubular lumen.


Subject(s)
Angiotensin II/pharmacology , Angiotensin II/urine , Benzimidazoles/pharmacology , Blood Pressure/drug effects , Kidney/drug effects , Receptor, Angiotensin, Type 1/metabolism , Tetrazoles/pharmacology , Analysis of Variance , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Biphenyl Compounds , Chromatography, Liquid , Hypertension, Renal/chemically induced , Hypertension, Renal/metabolism , Hypertension, Renal/physiopathology , Kidney/metabolism , Male , Radioimmunoassay , Rats , Rats, Sprague-Dawley , Renin-Angiotensin System/drug effects
18.
Am J Physiol Renal Physiol ; 298(1): F37-48, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19846569

ABSTRACT

Combination therapy of angiotensin-converting enzyme (ACE) inhibition and AT(1) receptor blockade has been shown to provide greater renoprotection than ACE inhibitor alone in human diabetic nephropathy, suggesting that ACE-independent pathways for ANG II formation are of major significance in disease progression. Studies were performed to determine the magnitude of intrarenal ACE-independent formation of ANG II in type II diabetes. Although renal cortical ACE protein activity [2.1 +/- 0.8 vs. 9.2 +/- 2.1 arbitrary fluorescence units (AFU) x mg(-1) x min(-1)] and intensity of immunohistochemical staining were significantly reduced and ACE2 protein activity (16.7 +/- 3.2 vs. 7.2 +/- 2.4 AFU x mg(-1) x min(-1)) and intensity elevated, kidney ANG I (113 +/- 24 vs. 110 +/- 45 fmol/g) and ANG II (1,017 +/- 165 vs. 788 +/- 99 fmol/g) levels were not different between diabetic and control mice. Afferent arteriole vasoconstriction due to conversion of ANG I to ANG II was similar in magnitude in kidneys of diabetic (-28 +/- 3% at 1 microM) and control (-23 +/- 3% at 1 microM) mice; a response completely inhibited by AT(1) receptor blockade. In control kidneys, afferent arteriole vasoconstriction produced by ANG I was significantly attenuated by ACE inhibition, but not by serine protease inhibition. In contrast, afferent arteriole vasoconstriction produced by intrarenal conversion of ANG I to ANG II was significantly attenuated by serine protease inhibition, but not by ACE inhibition in diabetic kidneys. In conclusion, there is a switch from ACE-dependent to serine protease-dependent ANG II formation in the type II diabetic kidney. Pharmacological targeting of these serine protease-dependent pathways may provide further protection from diabetic renal vascular disease.


Subject(s)
Angiotensin II/metabolism , Diabetes Mellitus, Type 2/metabolism , Kidney/metabolism , Peptidyl-Dipeptidase A/metabolism , Signal Transduction/physiology , Angiotensin I/metabolism , Angiotensin-Converting Enzyme 2 , Angiotensinogen/urine , Animals , Arterioles/pathology , Arterioles/physiopathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Diabetic Angiopathies/etiology , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/pathology , Disease Models, Animal , Kidney/blood supply , Male , Mice , Mice, Mutant Strains , Rats , Rats, Sprague-Dawley , Receptors, Leptin/genetics , Serine Proteases/metabolism , Vasoconstriction/physiology
19.
Hypertension ; 54(1): 120-6, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19487583

ABSTRACT

Chronic angiotensin II (Ang II) infusions enhance urinary excretion of angiotensinogen, suggesting augmentation of distal nephron sodium reabsorption. To assess whether chronic Ang II infusions (15 ng/min for 2 weeks) enhance distal nephron sodium reabsorption, we compared sodium excretion before and after blockade of the 2 main distal nephron sodium transporters by IV amiloride (5 mg/kg of body weight) plus bendroflumethiazide (12 mg/kg of body weight) in male C57/BL6 anesthetized control mice (n=10) and in chronic Ang II-infused mice (n=8). Chronic Ang II infusions increased systolic blood pressure to 141+/-6 mm Hg compared with 106+/-4 mm Hg in control mice. After anesthesia, mean arterial pressure averaged 97+/-4 mm Hg in chronic Ang II-infused mice compared with 94+/-3 mm Hg in control mice, allowing comparison of renal function at similar arterial pressures. Ang II-infused mice had lower urinary sodium excretion (0.16+/-0.04 versus 0.30+/-0.05 microEq/min; P<0.05), higher distal sodium reabsorption (1.74+/-0.18 versus 1.12+/-0.18 microEq/min; P<0.05), and higher fractional reabsorption of distal sodium delivery (91.1+/-1.8% versus 77.9+/-4.3%; P<0.05) than control mice. Urinary Ang II concentrations, measured during distal blockade, were greater in Ang II-infused mice (1235.0+/-277.2 versus 468.9+/-146.9 fmol/mL; P<0.05). In chronic Ang II-infused mice treated with spironolactone (n=5), fractional reabsorption of distal sodium delivery was similarly augmented as in chronic Ang II-infused mice (94.6+/-1.7%; P<0.01). These data provide in vivo evidence that there is enhanced distal sodium reabsorption dependent on sodium channel and Na(+)-Cl(-) cotransporter activity and increased urinary Ang II concentrations in mice infused chronically with Ang II.


Subject(s)
Angiotensin II/pharmacology , Nephrons/metabolism , Sodium/metabolism , Angiotensin II/administration & dosage , Angiotensin II/urine , Animals , Blood Pressure/drug effects , Glomerular Filtration Rate/drug effects , Infusion Pumps , Male , Mice , Mice, Inbred C57BL , Nephrons/blood supply , Renal Plasma Flow/drug effects , Sodium/urine , Urodynamics/drug effects , Vasoconstrictor Agents/administration & dosage , Vasoconstrictor Agents/pharmacology
20.
Am J Physiol Renal Physiol ; 296(5): F1067-71, 2009 May.
Article in English | MEDLINE | ID: mdl-19244403

ABSTRACT

In angiotensin II (ANG II)-induced hypertension, intrarenal ANG II levels are increased by AT(1) receptor-mediated ANG II internalization and endogenous ANG II generation. The objective of the present study was to determine the relative contribution of de novo formation of endogenous ANG II. Male Sprague-Dawley rats were divided into three groups: sham operated (n = 6), Val(5)-ANG II infused (n = 16), and Ile(5)-ANG II infused (n = 6). Val(5)-ANG II and Ile(5)-ANG II were infused at 80 ng/min via subcutaneous osmotic minipump for 13 days, followed by harvesting of blood and kidney samples. In six Val(5)-ANG II-infused rats, urine was collected on the day before infusion and on day 12 of infusion. Extracted samples were subjected to HPLC to separate Val(5)-ANG II from Ile(5)-ANG II followed by RIA. Systolic blood pressure increased significantly from 121 +/- 2 to 206 +/- 4 mmHg in the Val(5)-ANG II-infused rats and from 124 +/- 3 to 215 +/- 5 mmHg in the Ile(5)-ANG II-infused rats. In the Val(5)-ANG II-infused rats, the plasma Ile(5)-ANG II levels increased 196.2 +/- 70.1% compared with sham plasma Ile(5)-ANG II concentration. Val(5)-ANG II levels were 150.0 +/- 28.2 fmol/ml which accounted for 53.5 +/- 10.1% of the total ANG II in plasma. The kidney Ile(5)-ANG II levels in the Val(5)-ANG II-infused rats increased 69.9 +/- 30.7% compared with sham kidney Ile(5)-ANG II concentrations. Intrarenal accumulation of Val(5)-ANG II accounted for 52.5 +/- 5.3% of the total kidney ANG II during Val(5)-ANG II infusion while endogenous Ile(5)-ANG II accounted for 47.5 +/- 8.6%. The urinary Ile(5)-ANG II excretion rate on day 12 increased 93.2 +/- 32.1% compared with preinfusion level indicating increased formation of endogenous ANG II. Thus, the increases in intrarenal ANG II levels during chronic ANG II infusions involve substantial stimulation of endogenous ANG II formation which contributes to overall augmentation of intrarenal ANG II.


Subject(s)
Angiotensin II/metabolism , Angiotensin II/pharmacology , Hypertension, Renal/metabolism , Kidney/metabolism , Vasoconstrictor Agents/metabolism , Vasoconstrictor Agents/pharmacology , Angiotensin II/analogs & derivatives , Animals , Chromatography, High Pressure Liquid , Disease Models, Animal , Hypertension, Renal/chemically induced , Infusion Pumps, Implantable , Kidney/drug effects , Male , Rats , Rats, Sprague-Dawley , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...