Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 219
Filter
Add more filters










Publication year range
2.
Am J Physiol Heart Circ Physiol ; 298(6): H1807-18, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20363893

ABSTRACT

We have generated transgenic mice that express angiotensin II (ANG II) fused downstream of enhanced cyan fluorescent protein, expression of which is regulated by the mouse metallothionein promoter. The fusion protein, which lacks a secretory signal, is retained intracellularly. In the present study, RT-PCR, immunoblot analyses, whole-animal fluorescent imaging, and fluorescent microscopy of murine embryonic fibroblasts confirm expression of the fusion protein in vivo and in vitro. The transgene is expressed in all tissues tested (including brain, heart, kidney, liver, lung, and testes), and radioimmunoassay of plasma samples obtained from transgenic mice indicate no increase in circulating ANG II over wild-type levels, consistent with intracellular retention of the transgene product. Kidneys from transgenic and corresponding wild-type littermates were histologically evaluated, and abnormalities in transgenic mice consistent with thrombotic microangiopathy were observed; microthrombosis was frequently observed within the glomerular capillaries and small vessels. In addition, systolic and diastolic blood pressures, measured by telemetry (n = 8 for each group), were significantly higher in transgenic mice compared with wild-type littermates. Blood pressure of line A male transgenic mice was 125 + or - 1.7 over 97 + or - 1.6 compared with 109 + or - 1.7 over 83 + or - 1.4 mmHg in wild-type littermates (systolic over diastolic). In summary, overexpression of an intracellular fluorescent fusion protein of ANG II correlates with elevated blood pressure and kidney pathology. This transgenic model may be useful to further explore the intracellular renin-angiotensin system and its implication in abnormal kidney function and hypertension.


Subject(s)
Angiotensin II/metabolism , Green Fluorescent Proteins/metabolism , Hypertension/metabolism , Kidney/blood supply , Kidney/metabolism , Thrombotic Microangiopathies/metabolism , Angiotensin II/genetics , Animals , Blood Pressure/physiology , Cells, Cultured , Disease Models, Animal , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Green Fluorescent Proteins/genetics , Hypertension/pathology , Hypertension/physiopathology , Kidney/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/metabolism , Thrombotic Microangiopathies/pathology , Thrombotic Microangiopathies/physiopathology
3.
Acta Physiol Hung ; 96(3): 375-82, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19706379

ABSTRACT

This article pays tribute to the tremendous achievements of Dr. László Rosivall in renal (patho)physiology research and nephrology education in Hungary on the occasion of his 60th birthday. For the past several decades Dr. Rosivall has been a charismatic leader of academic institutions, national and international societies, foundations in physiology, nephrology and hypertension, but the most important of his many contributions, is his role as a scientist. He earned his MD with Summa cum Laude at Semmelweis University (1973) and was invited immediately after that to join the laboratory of Hársing. He studied the distribution of intra-renal blood flow employing then state-of-the-art methods as well as developed his own technique at Semmelweis University and at the University of Bergen with Knut Aukland. This led to his PhD thesis and degree in 1980. An important determinant of his early basic scientific training and development was his postdoctoral research fellowship and later many visiting professorships in the Nephrology Research and Training Center (NRTC) at the University of Alabama at Birmingham, Birmingham, AL, USA between 1981 and 1983. Actually, this research fellowship not only impacted his own future career, but it also cleared the path for many other young Hungarian scientists who later trained with Dr. Rosivall and then at UAB. The early 1980s were the years of significant scientific discoveries and the NRTC team at UAB made important contributions by their studies on renal and glomerular hemodynamics, the renin-angiotensin system (12, 19, 22) and by the development of classic experimental techniques like renal micropuncture, microperfusion, and the juxtamedullary nephron preparation (3) that are still being used worldwide. When Dr. Rosivall joined UAB in the 1980s, the team at the NRTC included Drs. Navar, Bell, Inscho, Carmines, Casellas, and Oparil, among many others, who share their fond memories of working with Dr. Rosivall in this article.


Subject(s)
Biomedical Research/history , Nephrology/history , History, 20th Century , Hungary , Nephrology/education
4.
Am J Physiol Renal Physiol ; 295(3): F772-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18579707

ABSTRACT

The objectives of this study were to determine the effects of chronic angiotensin II (ANG II) infusions on ANG II content and angiotensinogen expression in the mouse kidney and the role of the angiotensin II type 1 receptor (AT(1)R) in mediating these changes. C57BL/6J male mice were subjected to ANG II infusions at doses of 400 or 1,000 ng.kg(-1).min(-1) either alone or with an AT(1)R blocker (olmesartan; 3 mg.kg(-1).day(-1)) for 12 days. Systolic and mean arterial pressures were determined by tail-cuff plethysmography and radiotelemetry. On day 13, blood and kidneys were collected for ANG II determinations by radioimmunoanalysis and intrarenal angiotensinogen expression studies by quantitative RT-PCR, Western blotting, and immunohistochemistry. ANG II infusions at the low dose elicited progressive increases in systolic blood pressure (135 +/- 2.5 mmHg). In contrast, the high dose induced a rapid increase (152 +/- 2.5, P < 0.05 vs. controls, 109 +/- 2.8). Renal ANG II content was increased by ANG II infusions at the low dose (1,203 +/- 253 fmol/g) and the high dose (1,258 +/- 173) vs. controls (499 +/- 40, P < 0.05). Kidney angiotensinogen mRNA and protein were increased only by the low dose to 1.13 +/- 0.02 and 1.26 +/- 0.10, respectively, over controls (1.00, P < 0.05). These effects were not observed in mice infused at the high dose and those receiving olmesartan. The results indicate that chronic ANG II infusions augment mouse intrarenal ANG II content with AT(1)R-dependent uptake occurring at both doses, but only the low dose of infusion, which elicited a slow progressive response, causes an AT(1)R-dependent increase in intrarenal angiotensinogen expression.


Subject(s)
Angiotensin II/metabolism , Angiotensinogen/metabolism , Hypertension/metabolism , Kidney/metabolism , Receptor, Angiotensin, Type 1/metabolism , Angiotensin II/administration & dosage , Angiotensin II/blood , Animals , Blood Pressure , Body Weight , Hypertension/chemically induced , Hypertension/pathology , Kidney/pathology , Male , Mice , Mice, Inbred C57BL , Organ Size , Renin/blood , Renin/metabolism
5.
Cell Mol Biol (Noisy-le-grand) ; 53(4): 51-60, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17531161

ABSTRACT

The goal of this study was to characterize the impact of induction or inhibition of the heme-HO system on renal apoptosis in clipped and non-clipped kidneys from 2K1C hypertensive rats. Male Sprague-Dawley rats had a 0.25 mm silver clip placed around the left renal artery. Four groups of rats were studied: sham operated animals, 2K1C control rats, 2K1C rats received weekly injections of CoPP (5 mg/100 g body wt, administered subcutaneously), and 2K1C rats pretreated with SnMP (5 mg/ 100g body wt, administered intraperitoneally three times a week). The animals were sacrificed three weeks after surgery. We measured systolic blood pressure, plasma renin activity, non-clipped and clipped kidney HO-1 and HO-2 protein expression, HO activity, heme content, nitrotyrosine levels, and activation of selected pro- and anti-apoptotic proteins. Systolic blood pressure and plasma renin activity were significantly higher in 2K1C rats compared to sham rats. Compared to kidneys from sham animals, clipped kidneys from 2K1C rats showed a significant increase in HO-1 expression with increases in HO activity (26%), heme content (47%) and nitrotyrosine levels (49%), accompanied by an increase in caspase-3 and caspase-9 activity. In contrast, non-clipped kidneys from 2K1C rats showed no differences in HO-1 expression, HO activity, heme content, nitrotyrosine levels and caspase activity compared to sham rats. In clipped kidneys from 2K1C rats, inhibition of HO activity by SnMP augmented caspase-3 and caspase-9 activity and decreased expression of the anti-apoptotic Bcl-2 protein, while induction of HO-1 with CoPP strongly inhibited the activity of both caspases and increased the induction of Bcl-2 and Bcl-xl proteins. These findings demonstrate that the clipped kidneys responded to decreased renal perfusion pressure and increased oxidative stress by activation of the heme-HO system, which exerts antiapoptotic action via mechanisms involving decreased caspase-3 and caspase-9 activity, and increased expression of antiapoptotic molecules.


Subject(s)
Apoptosis/physiology , Heme Oxygenase-1/genetics , Hypertension, Renovascular/genetics , Animals , Caspases/metabolism , Gene Expression Regulation , Heme/analysis , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1/metabolism , Hypertension, Renovascular/enzymology , Hypertension, Renovascular/etiology , Hypertension, Renovascular/pathology , Kidney/chemistry , Kidney/enzymology , Kidney/pathology , Kidney/surgery , Male , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Tyrosine/analogs & derivatives , Tyrosine/analysis , bcl-X Protein/metabolism
6.
Am J Physiol Renal Physiol ; 293(2): F533-40, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17522266

ABSTRACT

During ischemia or hypoxia an increase in intracellular cytosolic Ca(2+) induces deleterious events but is also implicated in signaling processes triggered in such conditions. In MDCK cells (distal tubular origin), it was shown that mitochondria confer protection during metabolic inhibition (MI), by buffering the Ca(2+) overload via mitochondrial Na(+)-Ca(2+) exchanger (NCX). To further assess this process in cells of human origin, human cortical renal epithelial cells (proximal tubular origin) were subjected to MI and changes in cytosolic Ca(2+) ([Ca(2+)](i)), Na(+), and ATP concentrations were monitored. MI was accomplished with both antimycin A and 2-deoxyglucose and induced a 3.5-fold increase in [Ca(2+)](i), reaching 136.5 +/- 15.8 nM in the first 3.45 min. Subsequently [Ca(2+)](i) dropped and stabilized to 62.7 +/- 7.3 nM by 30 min. The first phase of the transient increase was La(3+) sensitive, not influenced by diltiazem, and abolished when mitochondria were deenergized with the protonophore carbonylcyanide p-trifluoromethoxyphenylhydrazone. The subsequent recovery phase was impaired in a Na(+)-free medium and weakened when the mitochondrial NCX was blocked with 7-chloro-5-(2-chlorophenyl)-1,5-dihydro-4,1-benzothiazepin-2(3H)-one (CGP-37157). Thus Ca(2+) entry is likely mediated by store-operated Ca(2+) channels and depends on energized mitochondria, whereas [Ca(2+)](i) recovery relied partially on the activity of mitochondrial NCX. These results indicate a possible mitochondrial-mediated signaling process triggered by MI, support the hypothesis that mitochondrial NCX has an important role in the Ca(2+) clearance, and overall suggest that mitochondria play a preponderant role in the regulation of responses to MI in human renal epithelial cells.


Subject(s)
Antimetabolites/pharmacology , Calcium Signaling/physiology , Kidney Tubules, Proximal/metabolism , Mitochondria/metabolism , Adenosine Triphosphate/metabolism , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Cell Line , Clonazepam/analogs & derivatives , Clonazepam/pharmacology , Humans , Kidney Tubules, Proximal/drug effects , Membrane Potentials/drug effects , Mitochondria/drug effects , Sodium/metabolism , Sodium-Calcium Exchanger/metabolism , Thiazepines/pharmacology , Uncoupling Agents/pharmacology
7.
Acta Physiol Hung ; 94(1-2): 31-48, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17444274

ABSTRACT

It is now well accepted that alterations in kidney function, due either to primary renal disease or to inappropriate hormonal influences on the kidney, are a cardinal characteristic in all forms of hypertension, and lead to a reduced ability of the kidneys to excrete sodium and the consequent development of elevated arterial pressures. However, it is also apparent that many extrarenal factors are important contributors to altered kidney function and hypertension. Central to many hypertensinogenic processes is the inappropriate activation of the renin-angiotensin system (RAS) and its downstream consequences by various pathophysiologic mechanisms. There may also be derangements in arachidonic acid metabolites, endothelium derived factors such as nitric oxide and carbon monoxide, and various paracrine and neural systems that normally interact with or provide a counteracting balance to the actions of the RAS. Thus, when the capacity of the kidneys to maintain sodium balance and extracellular fluid volume within appropriate ranges is compromised, increases in arterial pressure become necessary to re-establish normal balance.


Subject(s)
Angiotensin II/physiology , Hypertension/physiopathology , Renin-Angiotensin System/physiology , Animals , Atrial Natriuretic Factor/physiology , Blood Pressure/physiology , Carbon Monoxide/physiology , Cyclooxygenase Inhibitors/pharmacology , Eicosanoids/biosynthesis , Heme/metabolism , Heme Oxygenase (Decyclizing)/physiology , Hypertension, Renal/etiology
8.
Am J Physiol Renal Physiol ; 290(5): F1270-5, 2006 May.
Article in English | MEDLINE | ID: mdl-16380466

ABSTRACT

Megalin is a multiligand receptor heavily involved in protein endocytosis. We recently demonstrated that megalin binds and mediates internalization of ANG II. Although there is a strong structural resemblance between ANG II and ANG-(1-7), their physiological actions and their affinity for the angiotensin type 1 receptor (AT(1)R) are dissimilar. Therefore, the hypothesis of the present work was to test whether megalin binds and internalizes ANG-(1-7). The uptake of ANG-(1-7) was determined by exposure of confluent monolayers of BN/MSV cells (a model representative of the yolk sac epithelium) to fluorescently labeled ANG-(1-7) (100 nM) and measurement of the amount of cell-associated fluorescence after 4 h by flow cytometry. Anti-megalin antisera and an AT(1)R blocker (olmesartan) were used to interfere with uptake via megalin and the AT(1)R, respectively. ANG-(1-7) uptake was prevented by anti-megalin antisera (63%) to a higher degree than olmesartan (13%) (P < 0.001). In analysis by flow cytometry of binding experiments performed in brush-border membrane vesicles isolated from kidneys of CD-1 mice, anti-megalin antisera interfered with ANG-(1-7) binding more strongly than olmesartan (P < 0.05 against positive control). Interactions of megalin with ANG-(1-7) at a molecular level were studied by surface plasmon resonance, demonstrating that ANG-(1-7) binds megalin dose and time dependently and with an affinity similar to ANG II. These results show that the scavenger receptor megalin binds and internalizes ANG-(1-7).


Subject(s)
Angiotensin I/metabolism , Antihypertensive Agents/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/physiology , Peptide Fragments/metabolism , Cell Culture Techniques , Epithelium , Flow Cytometry , Humans , Kidney/physiology , Renin-Angiotensin System/physiology , Yolk Sac/cytology
9.
Am J Physiol Renal Physiol ; 288(2): F420-7, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15467006

ABSTRACT

Megalin is an abundant membrane protein heavily involved in receptor-mediated endocytosis. The major functions of megalin in vivo remain incompletely defined as megalin typically faces specialized milieus such as glomerular filtrate, airways, epididymal fluid, thyroid colloid, and yolk sac fluid, which lack many of its known ligands. In the course of studies on ANG II internalization, we were surprised when only part of the uptake of labeled ANG II into immortalized yolk sac cells (BN-16 cells) was blocked by specific peptide inhibitors and direct competitors of the angiotensin type 1 receptor. This led us to test if megalin was a receptor for ANG II. Four lines of direct evidence demonstrate that megalin and, to a lesser extent, its chaperone protein cubilin are receptors for ANG II. First, in BN-16 cells anti-megalin and anti-cubilin antisera interfere with ANG II uptake. Second, also in BN-16 cells, pure ANG II competes for uptake of a known megalin ligand. Third, in proximal tubule cell brush-border membrane vesicles extracted from mice, anti-megalin antisera interfere with ANG II binding. Fourth, purified megalin binds ANG II directly in surface plasmon resonance experiments. The finding that megalin is a receptor for ANG II suggests a major new function for the megalin pathway in vivo. These results also indicate that ANG II internalization in some tissues is megalin dependent and that megalin may play a role in regulating proximal tubule ANG II levels.


Subject(s)
Angiotensin II/metabolism , Kidney Tubules, Proximal/physiology , Low Density Lipoprotein Receptor-Related Protein-2/physiology , Vasoconstrictor Agents/metabolism , Animals , Mice , Polymerase Chain Reaction , Receptor, Angiotensin, Type 1/biosynthesis
11.
Am J Physiol Renal Physiol ; 281(4): F665-73, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11553513

ABSTRACT

We have studied cardiovascular and renal phenotypes in Npr1 (genetic determinant of natriuretic peptide receptor-A; NPRA) gene-disrupted mutant mouse model. The baseline systolic arterial pressure (SAP) in 0-copy mutant (-/-) mice (143 +/- 2 mmHg) was significantly higher than in 2-copy wild-type (+/+) animals (104 +/- 2 mmHg); however, the SAP in 1-copy heterozygotes (+/-) was at an intermediate value (120 +/- 4 mmHg). To determine whether Npr1 gene function affects the renin-angiotensin-aldosterone system (RAAS), we measured the components of RAAS in plasma, kidney, and adrenal gland of 0-copy, 1-copy, and 2-copy male mice. Newborn (2 days after the birth) 0-copy pups showed 2.5-fold higher intrarenal renin contents compared with 2-copy wild-type counterparts (0-copy 72 +/- 12 vs. 2-copy 30 +/- 7 microg ANG I. mg protein(-1). h(-1), respectively). The intrarenal ANG II level in 0-copy pups was also higher than in 2-copy controls (0-copy 33 +/- 5 vs. 2-copy 20 +/- 2 pg/mg protein, respectively). However, both young (3 wk) and adult (16 wk) 0-copy mutant mice showed a dramatic 50-80% reduction in plasma renin concentrations (PRCs) and in expression of renal renin message compared with 2-copy control animals. In contrast, the adrenal renin content and mRNA expression levels were 1.5- to 2-fold higher in 0-copy adult mice than in 2-copy animals. The results suggest that inhibition of renal and systemic RAAS is a compensatory response that prevents greater increases in elevated arterial pressures in adult NPRA null mutant mice. However, the greater renin and ANG II levels seen in 0-copy newborn pups provide evidence that the direct effect of NPRA activation on renin is an inhibitory response.


Subject(s)
Angiotensin II/blood , Receptors, Atrial Natriuretic Factor/genetics , Renin-Angiotensin System/physiology , Renin/blood , Aldosterone/blood , Animals , Atrial Natriuretic Factor/blood , Gene Expression/physiology , Genotype , Guanylate Cyclase/metabolism , Homeostasis/physiology , Kidney/anatomy & histology , Kidney/metabolism , Mice , Mice, Knockout , Organ Size , RNA, Messenger/analysis , Receptors, Atrial Natriuretic Factor/metabolism , Renin/genetics , Reverse Transcriptase Polymerase Chain Reaction
12.
Am J Hypertens ; 14(6 Pt 2): 74S-82S, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11411769

ABSTRACT

Experimental evidence has now been amassed to indicate that inhibition of nitric oxide (NO) synthase reduces total or regional renal blood flow by approximately 25 to 30% and markedly increases the renal vascular resistance, demonstrating that basal release of NO helps to maintain the relatively low vascular resistance that is characteristic for the kidney. It has been demonstrated that intraarterial administration of NO synthase inhibitors causes marked reductions in sodium excretion without changes in filtered load and suppressed the arterial pressure-induced natriuretic responses in the kidney. We also demonstrated that a constant rate infusion of a NO donor in dogs pretreated with a NOS inhibitor resulted in increases in sodium excretion but failed to restore the slope of the relation between arterial pressure and sodium excretion, suggesting that an alteration in intrarenal NO production rate during changes in arterial pressure is involved in the mediation of pressure natriuresis. Further experiments in dogs performed in our laboratory have confirmed that there is a direct relationship between changes in arterial pressure and intrarenal NO activity measured using NO-sensitive microelectrodes in the renal tissue. These arterial pressure-induced changes in intrarenal NO activity were seen positively correlated with the changes in urinary excretion rates of sodium. Collectively, these data suggest that acute changes in arterial pressure alter intrarenal NO production, which inhibits tubular sodium reabsorption to manifest the phenomenon of pressure natriuresis.


Subject(s)
Diuresis/physiology , Nitric Oxide/physiology , Renal Circulation/physiology , Animals , Feedback , Glomerular Filtration Rate/physiology , Hemodynamics/physiology , Homeostasis/physiology , Kidney/metabolism , Natriuresis/physiology , Vascular Resistance/physiology
13.
Hypertension ; 37(5): 1329-35, 2001 May.
Article in English | MEDLINE | ID: mdl-11358949

ABSTRACT

Chronic infusion of angiotensin (Ang) II leads to the development of hypertension and enhances intrarenal Ang II content to levels greater than can be explained from the circulating concentrations of the peptide. We previously reported that renal angiotensinogen (Ao) mRNA is enhanced in Ang II-dependent hypertension and may contribute to augmented intrarenal Ang II levels, but the Ao protein levels were not significantly increased. Because a high-salt diet (H/S) has been shown to suppress renal expression of Ao mRNA, we examined the effects of chronic Ang II infusion on kidney and liver Ao mRNA and protein levels in male Sprague-Dawley rats (n=12) maintained on an 8% salt diet. Ang II was administered via osmotic minipumps (40 ng/min) to 1 group (n=6) while the remaining rats were sham-operated. A H/S diet alone did not alter systolic blood pressure in sham animals (109+/-6 mm Hg at day 12); however, Ang II infusions to the H/S rats significantly increased systolic blood pressure (167+/-7 at day 12) and intrarenal Ang II content (459+/-107 fmol/g versus 270+/-42) despite a marked suppression of plasma renin activity (0.9+/-0.2 ng Ang I. mL(-1). h(-1) versus 2.8+/-1.3). Ang II infusions significantly increased kidney Ao mRNA compared with the H/S diet alone by 1.9+/-0.1-fold. Western blot analysis of kidney protein extracts showed that the Ang II-infused rats had increased kidney Ao protein levels compared with the H/S diet alone (1.9+/-0.1-fold). Liver Ao mRNA and protein and plasma Ao protein were also significantly increased by Ang II infusions. These data demonstrate the effects of Ang II infusion to stimulate Ao mRNA and protein. Thus, the augmented intrarenal Ang II in Ang II-dependent hypertension may result, in part, by a positive amplification mechanism to activate renal expression of AO:


Subject(s)
Angiotensin II/blood , Angiotensins/blood , Hypertension/blood , Angiotensin II/metabolism , Angiotensins/biosynthesis , Angiotensins/genetics , Animals , Blood Pressure , Blotting, Western , Body Weight , Diet , Disease Models, Animal , Hypertension/chemically induced , Hypertension/metabolism , Kidney/metabolism , Liver/metabolism , Male , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Renin/blood , Sodium, Dietary
14.
Hypertension ; 37(2 Pt 2): 753-9, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11230369

ABSTRACT

We recently demonstrated a direct relationship between autoregulation-related changes in renal vascular resistance (RVR) and renal interstitial ATP concentrations. To assess the possible role for extracellular ATP in the regulation of tubuloglomerular feedback (TGF)-mediated autoregulatory adjustments in RVR, renal interstitial ATP concentrations were measured with microdialysis probes in anesthetized dogs at different renal arterial pressures (RAPs) within the autoregulatory range during augmented and diminished activity of the TGF mechanism. Stepwise reductions in RAP from ambient pressure (129+/-3 mm Hg) to 102+/-2 mm Hg (step 1) and 75+/-1 mm Hg (step 2) resulted in significant decreases in ATP concentrations from 9.0+/-0.8 to 6.3+/-0.6 nmol/L in step 1 and to 4.2+/-0.5 nmol/L in step 2. Changes in RVR were highly correlated with changes in ATP concentrations (r=0.86, P<0.001, n=12). Acetazolamide (100 microgram. kg(-1). min(-1), n=6), which increases solute delivery to the macula densa, thus augmenting TGF activity, significantly decreased renal blood flow (RBF) by -16+/-2% and glomerular filtration rate (GFR) by -22+/-4% and increased ATP concentrations from 8.4+/-0.7 to 15.5+/-1.4 nmol/L. Although basal RBF and GFR levels were reduced by the acetazolamide infusion, autoregulation efficiency was maintained, and interstitial ATP concentrations were significantly decreased in response to reductions in RAP by -36+/-4% in step 1 and by -54+/-2% in step 2. The relationship between changes in RVR and interstitial ATP concentrations was preserved during acetazolamide treatment (r=0.80, P<0.01). Inhibition of the TGF mechanism by furosemide significantly increased RBF by 33+/-6% and GFR by 13+/-2% and decreased ATP concentrations from 8.9+/-1.4 to 5.0+/-0.8 nmol/L (n=6). Furosemide caused marked impairment of RBF and GFR autoregulatory efficiency (by -14+/-3% and -11+/-3% in step 1 and by -26+/-2% and -18+/-4% in step 2, respectively). In the furosemide-treated kidneys, interstitial ATP levels remained low and were not altered during reductions in RAP (4.7+/-0.7 nmol/L in step 1 and 4.7+/-0.8 nmol/L in step 2), and changes in RVR did not exhibit a correlation with changes in ATP concentrations (r=0.22, P=0.30). These data support the hypothesis that extracellular ATP contributes to autoregulatory adjustments in RVR that are mediated by changes in activity of the TGF mechanism.


Subject(s)
Adenosine Triphosphate/metabolism , Blood Pressure/physiology , Kidney Glomerulus/physiology , Kidney/metabolism , Acetazolamide/pharmacology , Adenosine/metabolism , Animals , Carbonic Anhydrase Inhibitors/pharmacology , Diuretics/pharmacology , Dogs , Extracellular Space/metabolism , Feedback , Furosemide/pharmacology , Glomerular Filtration Rate/drug effects , Homeostasis , Kidney Glomerulus/drug effects , Microdialysis , Regional Blood Flow/drug effects , Renal Artery/physiology , Vascular Resistance
15.
Circ Res ; 88(3): 347-51, 2001 Feb 16.
Article in English | MEDLINE | ID: mdl-11179204

ABSTRACT

A direct relationship between renal arterial pressure (RAP) and renal interstitial hydrostatic pressure (RIHP) has been shown under conditions of efficient renal blood flow autoregulation. Experiments were performed in six anesthetized dogs to evaluate whether these RIHP responses to changes in RAP were modified during nitric oxide (NO) inhibition with nitro-L-arginine (NLA) or after administration of NO donor agents. A microtip catheter transducer was placed underneath the renal capsule to measure RIHP. Stepwise reductions in RAP (140 to 80 mm Hg) during control conditions resulted in decreases in RIHP from its basal value of 4.7+/-1.1 mm Hg with a slope of 0.04+/-0.026 mm Hg. mm Hg(-)(1) along with decreases in urinary nitrate/nitrite excretion rate (U(NOx)V). Renal cortical and medullary blood flows, measured by laser-Doppler flowmetry, exhibited high autoregulatory efficiency over this RAP range. The changes in RIHP during alterations in RAP were positively correlated (r=0.743; P:<0.001) with the changes in U(NOx)V but not with cortical or medullary blood flow. NLA infusion decreased RIHP to 1.9+/-0.5 mm Hg and also reduced U(NOx)V from 1.8+/-0.2 to 0.9+/-0.01 nmol. min(-)(1). g(-)(1). Infusion of NO donors restored RIHP (4.3+/-0.9 mm Hg) and U(NOx)V (1.5+/-0.2 nmol. min(-)(1). g(-)(1)). During NLA infusion, the RIHP responses to reductions in RAP were markedly attenuated and were not restored even during constant-rate infusion of NO donors. The results suggest that changes in RIHP in response to alterations in RAP are associated with changes in intrarenal NO, suggesting a direct effect of NO to regulate RIHP.


Subject(s)
Blood Pressure/physiology , Kidney/physiology , Nitric Oxide/physiology , Renal Artery/physiology , Animals , Blood Pressure/drug effects , Dogs , Enzyme Inhibitors/pharmacology , Hydrostatic Pressure , Kidney/blood supply , Kidney/drug effects , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitroarginine/pharmacology , Renal Artery/drug effects , Renal Circulation/drug effects
16.
Am J Physiol Renal Physiol ; 280(3): F406-14, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11181402

ABSTRACT

Adenosine vasoconstricts preglomerular arterioles via adenosine A1 receptors. Because adenosine also activates adenosine A2 receptors, its overall renal vascular actions are complex and not fully understood. The present study was performed to determine the relative contributions of adenosine A1 and A2a receptors to the responsiveness of the renal microvasculature to adenosine. Afferent and efferent arteriolar diameters were monitored in vitro using the blood-perfused rat juxtamedullary nephron preparation. Basal afferent and efferent arteriolar diameters averaged 17.1 +/- 0.5 (n = 35) and 17.8 +/- 0.5 (n = 20) microm, respectively. Superfusion with 0.1 and 1 micromol/l adenosine did not significantly alter afferent and efferent arteriolar diameters; however, 10 micromol/l adenosine significantly reduced afferent and efferent arteriolar diameters (-8.2 +/- 0.8 and -5.7 +/- 0.6%, respectively). The afferent and efferent arteriolar vasoconstrictor responses to adenosine waned at a dose of 100 micromol/l, such that diameters returned to values not significantly different from control within 2 min. During adenosine A1 receptor blockade with 8-noradamantan-3-yl-1,3-dipropylxanthine (KW-3902: 10 micromol/l), 10 and 100 micromol/l adenosine significantly increased afferent diameter by, respectively, 8.1 +/- 1.2 and 13.7 +/- 1.3% (n = 14) and efferent arteriolar diameter by 6.4 +/- 1.3 and 9.3 +/- 1.2% (n = 8). The afferent and efferent arteriolar vasodilatory responses to adenosine in the presence of KW-3902 were significantly attenuated by addition of the adenosine A2a receptor antagonist 1,3-dipropyl-7-methyl-8-(3,4-dimethoxystyryl)xanthine (KF-17837: 15 micromol/l, n = 7 and 6, respectively). The addition of KF-17837 alone significantly enhanced afferent (n = 15) and efferent (n = 6) arteriolar vasoconstrictor responses to 1, 10, and 100 micromol/l adenosine. These results indicate the presence of adenosine A1 and A2a receptors on afferent and efferent arterioles of juxtamedullary nephrons, such that adenosine A2a receptor-mediated vasodilation partially buffers adenosine-induced vasoconstriction in both pre- and postglomerular segments of the renal microvasculature.


Subject(s)
Receptors, Purinergic P1/physiology , Renal Circulation/physiology , Adenosine/metabolism , Adenosine/pharmacology , Animals , Arterioles/drug effects , Arterioles/physiology , Extracellular Space/metabolism , Male , Microcirculation/physiology , Purinergic P1 Receptor Antagonists , Rats , Rats, Sprague-Dawley , Receptor, Adenosine A2A , Vasomotor System/physiology , Xanthines/pharmacology
17.
Kidney Int ; 58(6): 2320-30, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11115066

ABSTRACT

BACKGROUND: Enhanced intrarenal angiotensin II (Ang II) activity may contribute to diabetic nephropathy. The proximal tubule is a proposed site of significant intrarenal Ang II production. We determined the effect of early diabetes on mRNA expression of components of the proximal tubule renin-angiotensin system. METHODS: Three groups of male Sprague-Dawley rats were studied after two weeks: (1) control (C), (2) streptozotocin-induced diabetes (STZ), and (3) STZ-induced diabetes, with normoglycemia maintained by insulin implants (STZ-I). Competitive reverse transcription-polymerase chain reaction was used to assay mRNA for renin, angiotensinogen, and angiotensin-converting enzyme in suspensions of proximal tubules; plasma and kidney levels of Ang II were measured by radioimmunoassay, and Western analysis of Ang II subtype 1 (AT1) receptors was performed. RESULTS: STZ rats tended to have increased plasma and intrarenal levels of Ang II compared with C and STZ-I rats. In proximal tubules, mRNA for renin was significantly increased in STZ rats, with reversal to control values in STZ-I rats (C, 2432 +/- 437 vs. STZ, 5688 +/- 890 fg/0.25 microg RNA, P < 0.05 vs. C, N = 9, vs. STZ-I, 1676 +/- 376 fg/0.25 microg RNA, P = NS vs. C). In STZ rats, the AT1 receptor antagonist losartan caused a further fivefold increase in proximal tubule renin mRNA, associated with proximal tubular renin immunostaining. STZ had no significant effect on mRNA expression for angiotensinogen or angiotensin-converting enzyme in proximal tubules. By Western blot analysis, cortical and proximal tubule AT1 receptor protein expression was significantly decreased in STZ rats. CONCLUSIONS: These data suggest activation of the proximal tubule renin-angiotensin system in early STZ diabetes, mediated at least partly by enhanced expression of renin mRNA. Increased local production of Ang II could contribute to tubulointerstitial injury in this model.


Subject(s)
Diabetic Nephropathies/physiopathology , Kidney Tubules, Proximal/physiopathology , Renin/genetics , Angiotensin II/analysis , Angiotensin II/blood , Angiotensin Receptor Antagonists , Angiotensinogen/genetics , Animals , Antihypertensive Agents/pharmacology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Gene Expression/physiology , Hypertrophy , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Kidney Tubules, Proximal/chemistry , Kidney Tubules, Proximal/pathology , Losartan/pharmacology , Male , Nephritis, Interstitial/genetics , Nephritis, Interstitial/pathology , Nephritis, Interstitial/physiopathology , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/genetics , Renin/analysis , Renin-Angiotensin System/physiology
18.
Am J Physiol Renal Physiol ; 279(5): F858-65, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11053046

ABSTRACT

The dynamic activity of afferent arteriolar diameter (AAD) and blood flow (AABF) responses to a rapid step increase in renal arterial pressure (100-148 mmHg) was examined in the kidneys of normal Sprague-Dawley rats (n = 11) before [tubuloglomerular feedback (TGF)-intact] and after interruption of distal tubular flow (TGF-independent). Utilizing the in vitro blood-perfused juxtamedullary nephron preparation, fluctuations in AAD and erythrocyte velocity were sampled by using analog-to-digital computerized conversion, video microscopy, image shearing, and fast-frame, slow-frame techniques. These assessments enabled dynamic characterization of the autonomous actions and collective interactions between the myogenic and TGF mechanisms at the level of the afferent arteriole. The TGF-intact and TGF-independent systems exhibited common initial (0-24 vs. 0-13 s, respectively) response slope kinetics (-0.53 vs. -0.47% DeltaAAD/s; respectively) yet different maximum vasoconstrictive magnitude (-11.28 +/- 0.1 vs. -7. 02 +/- 0.9% DeltaAAD; P < 0.05, respectively). The initial AABF responses similarly exhibited similar kinetics but differing magnitudes. In contrast, during the sustained pressure input (13-97 s), the maximum vasoconstrictor magnitude (-7.02 +/- 0.9% DeltaAAD) and kinetics (-0.01% DeltaAAD/s) of the TGF-independent system were markedly blunted whereas the TGF-intact system exhibited continued vasoconstriction with slower kinetics (-0.20% DeltaAAD/s) until a steady-state plateau was reached (-25.9 +/- 0.4% DeltaAAD). Thus the TGF mechanism plays a role in both direct mediation of vasoconstriction and in modulation of the myogenic response.


Subject(s)
Arterioles/physiology , Kidney Glomerulus/blood supply , Kidney Tubules/blood supply , Renal Circulation/physiology , Animals , Blood Flow Velocity/physiology , Blood Pressure/physiology , Feedback/physiology , In Vitro Techniques , Kidney Glomerulus/physiology , Kidney Tubules/physiology , Male , Models, Cardiovascular , Nephrons/blood supply , Nephrons/physiology , Rats , Rats, Sprague-Dawley , Regression Analysis , Vascular Patency/physiology , Vasoconstriction/physiology
19.
Am J Physiol Renal Physiol ; 279(2): F289-93, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10919848

ABSTRACT

The relative contributions of ANG II and bradykinin (BK) to the renal regional blood flow responses during angiotensin-converting enzyme (ACE) inhibition remain unclear. This study was performed to evaluate renal cortical (CBF) and medullary blood flow (MBF) responses to intrarterial administration of enalaprilat (33 microg. kg(-1). min (-1)) after blockade of the ANG II AT(1 )receptors with candesartan (100 microg) in 7 dogs fed a low-salt diet (0.01%) for 5 days. Laser-Doppler flowmetry was used to measure relative changes in CBF and MBF. Candesartan alone increased CBF (+20 +/- 2%) and MBF (+22 +/- 7%). Enalaprilat infusion after candesartan administration resulted in further increases in both CBF (+21 +/- 5%) and MBF (+41 +/- 8%). However, the relative changes in MBF were significantly greater (P < 0.01) than those in CBF. Administration of the BK B(2) receptor blocker icatibant (300 microg) after enalaprilat returned CBF and MBF to values seen with candesartan alone. These data support a substantive role for BK potentiation during ACE inhibitor-induced renal vasodilation in dogs maintained on a low-sodium diet, with a relatively greater effect on MBF compared to CBF.


Subject(s)
Angiotensin II/physiology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Bradykinin/physiology , Renal Circulation/drug effects , Sodium/deficiency , Adrenergic beta-Antagonists/pharmacology , Angiotensin Receptor Antagonists , Animals , Benzimidazoles/pharmacology , Biphenyl Compounds , Bradykinin/analogs & derivatives , Bradykinin/pharmacology , Diet, Sodium-Restricted , Dogs , Laser-Doppler Flowmetry , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Tetrazoles/pharmacology
20.
Am J Physiol Renal Physiol ; 279(2): F319-25, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10919852

ABSTRACT

Chronic infusions of initially subpressor doses of angiotensin II (ANG II) lead to progressive hypertension over a 2-wk period and to augmented intrarenal ANG II levels. The present study was performed to investigate total renal blood flow (RBF) and medullary blood flow (MBF) autoregulatory behavior and pressure-natriuresis in ANG II-infused hypertensive rats and how these are modified by concomitant treatment with an ANG II AT(1) receptor antagonist. ANG II-infused rats (n = 27) were prepared by administration of ANG II at 60 ng/min via osmotic minipump for 13 days. Twelve of the ANG II-infused hypertensive rats were treated with losartan in the drinking water (30 mg. kg.(-1) day(-1)). Rats were anesthetized with pentobarbital sodium (50 mg/kg, ip) and prepared for renal function measurements. An aortic clamp was placed above the junction of the left renal artery to reduce renal arterial pressure. Autoregulatory responses for renal plasma flow, overall RBF, and glomerular filtration rate were impaired in ANG II-infused hypertensive rats; however, MBF autoregulation was not disrupted. Most strikingly, pressure-natriuresis was markedly suppressed in ANG II-infused hypertensive rats. Chronic treatment with losartan prevented the impairment of the pressure-natriuresis relationship caused by chronic ANG II infusion. These findings demonstrate that chronic ANG II infusion leads to marked impairment of sodium excretion and suppression of the pressure-natriuresis relationship, which may contribute to the progressive hypertension that occurs in this model. These renal effects are prevented by simultaneous treatment with an AT(1) receptor blocker.


Subject(s)
Angiotensin II/pharmacology , Blood Pressure , Homeostasis , Hypertension/physiopathology , Kidney/physiopathology , Natriuresis , Animals , Diuresis , Glomerular Filtration Rate , Hypertension/urine , Kidney Medulla/blood supply , Male , Rats , Rats, Inbred SHR/physiology , Rats, Sprague-Dawley , Renal Circulation
SELECTION OF CITATIONS
SEARCH DETAIL
...