Departments of Cellular and Molecular Medicine and Medicine, Division of Nephrology, The Kidney Research Centre, Ottawa Hospital Research Institute and University of Ottawa, Ottawa, Ontario, Canada K1H 8L6; and the Department of Medicine, University of Virginia Health System, Charlottesville, Virginia 22908
![]() |
ABSTRACT |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
The interaction of ANG II with intrarenal AT1 receptors has been implicated in the progression of diabetic nephropathy, but the role of intrarenal AT2 receptors is unknown. The present studies determined the effect of early diabetes on components of the glomerular renin-angiotensin system and on expression of kidney AT2 receptors. Three groups of rats were studied after 2 wk: 1) control (C), 2) streptozotocin (STZ)-induced diabetic (D), and 3) STZ-induced diabetic with insulin implant (D+I), to maintain normoglycemia. By competitive RT-PCR, early diabetes had no significant effect on glomerular mRNA expression for renin, angiotensinogen, or angiotensin-converting enzyme (ACE). In isolated glomeruli, nonglycosylated (41-kDa) AT1 receptor protein expression (AT1A and AT1B) was increased in D rats, with no change in glycosylated (53-kDa) AT1 receptor protein or in AT1 receptor mRNA. By contrast, STZ diabetes caused a significant decrease in glomerular AT2 receptor protein expression (47.0 ± 6.5% of C; P < 0.001; n = 6), with partial reversal in D+I rats. In normal rat kidney, AT2 receptor immunostaining was localized to glomerular endothelial cells and tubular epithelial cells in the cortex, interstitial, and tubular cells in the outer medulla, and inner medullary collecting duct cells. STZ diabetes caused a significant decrease in AT2 receptor immunostaining in all kidney regions, an effect partially reversed in D+I rats. In summary, early diabetes has no effect on glomerular mRNA expression for renin, angiotensinogen, or ACE. AT2 receptors are present in glomeruli and are downregulated in early diabetes, as are all kidney AT2 receptors. Our data suggest that alterations in the balance of kidney AT1 and AT2 receptor expression may contribute to ANG II-mediated glomerular injury in progressive diabetic nephropathy.
renin-angiotensin system; glomerulus; immunohistochemistry; hyperglycemia; angiotensin type 1 receptor
![]() |
INTRODUCTION |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
ANG II IS synthesized within the kidney and is a mediator of progressive injury in diabetic nephropathy. Although evidence suggests activation of the intrarenal renin-angiotensin system (RAS) in diabetes (23), the source of intrarenal ANG II formation remains unclear. Anderson et al. (3) demonstrated an increase in kidney renin mRNA and activity in early streptozotocin (STZ)-induced diabetes in rats, and a stimulatory effect of high glucose on proximal tubule angiotensinogen expression was found in cell culture studies (42). In rats with early STZ diabetes, we recently demonstrated increased proximal tubule cell mRNA expression for renin, with reversal by insulin, suggesting that hyperglycemia may selectively augment proximal tubule ANG II synthesis (51).
The glomerulus also has the capacity for synthesis of ANG II (5). Components of the renin-angiotensin system have been detected in cultured rat (8) and human (19) mesangial cells, and glomerular endothelial cells express angiotensin-converting enzyme (ACE) (46). The effect of diabetes on expression of components of the glomerular RAS has not been extensively studied, although glomerular ACE appears to be upregulated in early diabetes in both rats (3) and humans (25).
Once ANG II is formed in the kidney, it appears to
exert most of its hemodynamic and nonhemodynamic effects via
interaction with plasma membrane AT1 receptors.
AT1 receptor stimulation causes enhanced vasoconstriction
of the efferent arteriole compared with the afferent arteriole
(48), which may contribute to increased glomerular
capillary pressure and mechanical stretch-induced glomerular injury
(2). In addition, ANG II binds to AT1
receptors present on glomerular mesangial cells, tubular cells, and
interstitial cells, resulting in cell hypertrophy, and is associated
with stimulation of extracellular matrix production and elaboration of
the profibrotic cytokine transforming growth factor- (TGF-
)
(15, 44). A number of studies have demonstrated a
downregulation of glomerular and tubular AT1 receptors in
early diabetes (7, 9, 11, 16, 43). Nonetheless, the
importance of AT1 receptor activation in mediating
progressive diabetic glomerulosclerosis is suggested by studies in
experimental diabetes in which AT1 receptor blockade significantly reduced proteinuria and inhibited development of glomerulosclerosis (29, 38), similar to the beneficial
effects of ACE inhibitors in diabetic humans (1, 19, 28,
37) and animals (38, 49).
ANG II AT2 receptors have recently been detected in adult rat kidney by immunohistochemistry (24, 27). In contrast to AT1 receptors, AT2 receptors are linked to reduction of blood pressure (14, 35), inhibition of cell growth (22, 26), and apoptosis (47). In glomerular endothelial cells, AT2 receptors stimulate expression of the chemokine regulated on activation of normal T cells expressed and secreted (RANTES), which may contribute to recruitment of monocytes and macrophages in glomerular inflammation (45). The role or regulation of AT2 receptors in diabetic nephropathy, however, is unknown.
In the present studies, we determined the effect of early diabetes in rats on mRNA expression of components of the glomerular RAS, and on glomerular AT1 and AT2 receptor expression. We also performed immunohistochemistry to localize AT2 receptor expression in the normal and diabetic kidney. Our results indicate that diabetes has no significant effect on expression of mRNA for glomerular renin, angiotensinogen, ACE, or AT1 receptors but decreases glomerular AT2 receptors. Furthermore, AT2 receptor expression is decreased in all kidney regions in early diabetes. The data suggest that decreased AT2 receptor expression in the diabetic kidney may be a determinant of the rate of progression of glomerular injury.
![]() |
METHODS |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Animal model. Age-matched male Sprague-Dawley rats, weighing 200-225 g, were used for animal studies, after acclimatization for 5 days. Animals were allowed free access to distilled water and standard rat chow. All studies were approved by the Animal Care Committee of the University of Ottawa.
Rats were rendered diabetic with STZ (65 mg/kg ip; Sigma, St. Louis, MO) dissolved in 0.1 M sodium citrate buffer (pH 4.0). After 24 h, urine was assessed for glucose and ketones with a Keto-Diastix reagent strip (Bayer, Etobicoke, ON), and only those animals with sustained glucosuria were classified as diabetic and were included in further experiments. Rats were divided into three groups: 1) control (C) rats, injected with vehicle (0.1 M sodium citrate buffer, pH 4.0); 2) diabetic (D), rats treated daily with 1-2 units of insulin (Humulin L, Eli Lilly, Indianapolis, IN) subcutaneously to maintain hyperglycemia but prevent ketosis; and 3) diabetic plus insulin implant (D+I) rats, implanted with a sustained-release insulin implant (Linplant, Linshin Canada, Scarborough, ON), to maintain euglycemia. After 2 wk, rats were killed by CO2 narcosis followed by decapitation, and blood was collected for glucose, creatinine, and renin activity analysis. Measurements were performed by the Ottawa Hospital Biochemistry Laboratory (General Campus).Isolation of glomeruli. Glomeruli were isolated by Percoll gradient centrifugation, modified from a previously described method (39). Renal cortexes from both rat kidneys were dissected, and gently minced in a glass petri dish on ice. The tissue was then suspended in a solution containing (in mM) 115 NaCl, 24 NaCO3, 5 KCl, 1.5 CaCl2, 1.0 MgSO4, 2.0 NaH2PO4, 5.0 glucose, 1.0 alanine, and 10.0 HEPES, pH 7.4, as well as 0.03% collagenase (type IV; Sigma) and 0.01% soybean trypsin inhibitor (Sigma; buffer A). The suspension was gassed with 95% O2-5% CO2 and placed in a 37°C water bath for 45 min. After digestion, the cortical suspension was strained through a 250-µm brass sieve (mesh no. 60, Newark Wire Cloth, ESBE Scientific, Markham, ON) and centrifuged for 1 min at 100 g. The pellet was resuspended in buffer A without collagenase or trypsin inhibitor and centrifuged for 1 min. This was repeated three times. Next, the pellet was resuspended in a 40% Percoll (Sigma) solution of identical ionic composition as buffer A, which had been chilled to 4°C. The Percoll solution was centrifuged at 26,000 g for 30 min at 4°C, and the digested tissue was separated into four distinct bands (F1-F4) after centrifugation, as described (39). The uppermost band of the Percoll gradient (F1 layer) was highly enriched in glomeruli, as described (6). Subsequently, further purification of this band by multiple sieving was performed as described (5). Briefly, the F1 layer was removed and washed with PBS buffer (in mM): 8.5 Na2HPO4, 1.7 NaH2PO4, 145 NaCl, pH 7.4. The tissue was then strained through a 106-µm brass sieve (mesh no. 150) with PBS buffer, and glomeruli were collected on a 75-µm brass sieve (mesh no. 200) immediately below it. The collected tissue consisted of a pure (100%) collection of glomeruli, as determined by light microscopy. Glomerular cells did not undergo significant cell death in this preparation as determined by their ability (> 95%) to exclude the vital dye Trypan blue (10 mg/dl) (data not shown).
Competitive RT-PCR. Total RNA was isolated from glomeruli by using a commercial kit (RNeasy, Qiagen, Chatsworth, CA). RNA quality was assessed by running samples on ethidium bromide-stained 2% agarose-formaldehyde gels and by measuring optical density at 260 and 280 nm. RNA yield varied between 5 and 25 µg per glomerular isolation, and all samples were of high quality as assessed by these standards.
To measure absolute mRNA levels for renin, angiotensinogen, and ACE, a competitive RT-PCR assay was performed on total glomerular RNA by using deletion mutant cRNA for each of these components. Each sample of RNA (62.5 ng) was simultaneously reverse-transcribed with serial dilutions of deletion mutant cRNA, generated by in vitro transcription from plasmids containing mutant cDNA sequences. To generate deletion mutant cDNA, an inverse PCR method was utilized, essentially as described (12). Briefly, partial cDNA sequences for renin, angiotensinogen, and ACE were PCR amplified from rat kidney RNA and ligated into the pCR-Script SK(+) cloning vector (Stratagene, La Jolla, CA). The renin cDNA PCR product was generated with a sense primer, 5'-CTGCCACCTTGTTGTGTGAG-3', and an antisense primer, 5'-CCAGTATGCACAGGTCATCG-3', and corresponded to bases 1033-1296 of the rat renin cDNA (264-bp PCR product) (12). The angiotensinogen cDNA PCR product was generated with the sense primer 5'-CCTCGCTCTCTGGACTTATC-3' and the antisense primer 5'-CAGACACTGAGGTGCTGTTG-3' and corresponded to bases 737-962 of the rat angiotensinogen cDNA (226-bp PCR product) (12). Finally, the ACE cDNA PCR product was generated with the sense primer 5'-GCCACATCCAGTATTTCATGCAGT-3' and the antisense primer 5'-AACTGGAACTGGATGATGAAGCTGA-3' and corresponded to bases 3013-3454 of the rat ACE cDNA (442-bp PCR product) (17). All primers were obtained from Oligos Etc. (Wilsonville, OR). Inverse PCR was performed on the plasmids containing the renin, angiotensinogen, and ACE-partial cDNA sequences, by using sense and antisense primers oriented in a "tail-to-tail" direction, thereby amplifying the cloning vector and a fragment of the cDNA, with a gap between the 5' ends of the PCR product (12). The oligonucleotide primers for inverse PCR of the renin cDNA were sense, 5'-CGACTGAGCGTTGTGAACTGTAGCCA-3', corresponding to bases 1166-1183 of the rat renin cDNA, and antisense, 5'-CGACTGAGATATAGGATGTGCCAGTG-3', corresponding to bases 1093-1076, and resulting in a 208-bp deletion mutant product (deletion of 56 bp). For angiotensinogen, the sense primer, 5'-AGCGTCGTTCCAAGGGAAGATGAGAGGC-3', corresponded to bases 847-864 of the rat angiotensinogen cDNA, and the antisense primer was 5'-CGACTGAGTGTCACAGCCTGCACAAACC-3', corresponding to bases 811-791, resulting in a 169-bp deletion mutant PCR product (deletion of 57 bp). For ACE, the sense primer was 5'-GTCTCTGCCCTCCAGTGCCTAG-3', corresponding to bases 3337-3358 of the rat ACE cDNA, and the antisense primer was 5'-GAAAGTTGATGTCATGCTC-3', corresponding to bases 3195-3177, resulting in a 301-bp deletion mutant cDNA product (deletion of 141 bp). All inverse PCR reactions were performed for 35 cycles in a PerkinElmer Gene Amp 2400 PCR thermocycler, with hot-start at 96°C for 8 min, followed by denaturation at 96°C for 60 s, annealing at 63°C for 60 s, and elongation at 72°C for 90 s. Self-ligation of the PCR product was performed and plasmids were grown in competent Escherichia coli, followed by plasmid isolation (Qiagen). To confirm correct sequences and orientation, all deletion mutants were sequenced by the University of Ottawa DNA sequencing facility. Renin, angiotensinogen, and ACE cRNAs were transcribed from deletion mutant plasmids by using T3 or T7 RNA polymerase (Stratagene). The template was then degraded with amplification grade deoxyribonuclease I (DNase I, Life Technologies, Burlington, ON), and the RNA products were purified by phenol/chloroform extraction and ethanol precipitation. RNA was quantified by absorbance at 260 nm. For competitive RT-PCR reactions, RNA samples were treated with DNase I before reverse transcription, to digest residual genomic DNA. Samples of RNA (total RNA and dilutions of deletion mutant cRNA for either renin, angiotensinogen, or ACE) were then reverse-transcribed by using random hexamers (2.5 µM) and murine leukemia RT (2.5 U/µl) (Gene Amp RNA PCR kit, PerkinElmer, Branchburg, NJ). To control for possible genomic or plasmid DNA contamination, all experiments included a reaction in which RT was omitted from the transcription buffer. After reverse transcription, the cDNA mixture was amplified by PCR, in a total volume of 100 µl containing 2.5 U AmpliTaq DNA polymerase, 2 mM MgCl2, 1× PCR buffer II (PerkinElmer), and 1 µM each of the sense and antisense oligonucleotide PCR primers for the cDNA of interest. The sense and antisense primers for renin, angiotensinogen, and ACE were identical to those used to generate the nonmutant PCR products described above, of 264-, 226-, and 442-bp sizes, respectively. Preliminary experiments revealed that the yield of PCR products for all three components was linear up to 40 cycles of PCR. Accordingly, PCR was performed for 35 cycles, with a hot-start at 96°C for 3 min, followed by cycles at 94°C for 30 s, 63°C for 30 s, and 72°C for 45 s, followed by extension at 72°C for 10 min. To quantitate PCR products, samples were run on 3% agarose gels stained with Vistra green nucleic acid gel stain (Amersham). To determine the amounts of initial mRNA for renin, angiotensinogen, or ACE, PCR products and their corresponding deletion mutants were quantified by PhosphorImager analysis (Storm 860, Molecular Dynamics, Sunnyvale, CA). The logarithms of the ratio of target to deletion mutant (competitor) species were plotted as a function of the initial amount of deletion mutant cRNA. The initial amount of target mRNA was quantitated by extrapolation. Experiments were eliminated from further analysis if the correlation of fit (r2) for the generated curve was <0.9. To establish that the competitive RT-PCR assay could detect predicted changes in mRNA expression, normal rats were placed on diets consisting of either 2.9% NaCl (high salt or HS) or 0% NaCl (zero salt or ZS) for 5 days, because sodium depletion is known to stimulate glomerular renin mRNA expression (36). Competitive RT-PCR for renin mRNA was performed on total RNA isolated from cortex and glomeruli. Figure 1A illustrates a representative experiment in which 62.5 ng of total glomerular RNA were reverse transcribed and coamplified with serial dilutions of mutant renin cRNA, with renin primers. The ZS diet caused an approximately threefold increase in renin mRNA in cortex (Fig. 1B: HS: 925.2 ± 54.7 fg mRNA/62.5 ng total RNA vs. ZS: 2,773.7 ± 348.8 fg mRNA/62.5 ng total RNA; P < 0.001; n = 6), and glomeruli (HS: 2,241.0 ± 582.2 fg mRNA/62.5 ng total RNA vs. ZS: 6,223.8 ± 1,320.5 fg mRNA/62.5 ng total RNA; P < 0.025; n = 5).
|
Northern analysis for glomerular AT1 receptor mRNA.
Northern hybridization for AT1 receptor mRNA in glomeruli
was performed by using a 1.2-kb Sac I-Kpn I cDNA
fragment encoding the rat AT1A receptor, essentially as we
have previously performed (18). Briefly, total RNA was
isolated from glomeruli by using the RNeasy kit (Qiagen), and 5-µg
samples were run on 1% agarose-2.2 M formaldehyde gels and transferred
onto nylon membranes (Schleicher and Schuell, Keene, NH), followed by
ultraviolet crosslinking (Bio-Rad UV Gene Linker; Bio-Rad, Montreal,
QC). The rat AT1A cDNA probe, which hybridizes to both
AT1A and AT1B receptor mRNA, was labeled with
[32P]dCTP (3,000 Ci/mmol; Amersham) by the random primer
method (Multiprime DNA labeling system, Amersham). Hybridization was
performed overnight at 42°C, as described (18), and the
membranes were washed at low stringency (2× SSC, 0.1% SDS) where SSC
is standard sodium citrate, for 30 min at 23°C, and then at high
stringency (0.2× SSC, 0.1% SDS) for 15 min at 65°C. Membranes were
then exposed for 48 h at 70°C to Kodak Biomax MS film, with
two Cronex intensifiers (Sigma). After quantitation of the
AT1 receptor mRNA signal by densitometry, the membranes
were stripped and reprobed with [32P]dCTP-labeled cDNA
probe for human
-actin, as we have performed (10).
Immunohistochemistry. Kidneys were removed, cut longitudinally, and immediately placed in Zamboni's fixative (2% paraformaldehyde, 15% picric acid in PBS) for 2 h. The solution was replaced with fresh fixative and incubated at 4°C overnight. The following day the tissue was washed with 10% sucrose in PBS. The sucrose phosphate buffer was replaced daily on each of the following 7 days. Kidneys were then paraffin embedded, and 10-µm sections were cut. Prior to staining, sections were deparaffinized, and endogenous peroxidase activity was blocked by incubating the slides in 0.3% H2O2 in 100% MeOH for 30 min. To block nonspecific binding, sections were incubated in PBS containing 1% milk and 3% goat serum. Sections were then incubated for 48 h at 4°C in a humidified chamber with a rabbit anti-rat polyclonal AT2 receptor antibody (24) diluted 1:100 in PBS containing 1.5% goat serum and 0.5% milk. This antibody has a high degree of specificity for the rat AT2 receptor and has been used to detect AT2 receptors in the rat adrenal gland, rat brain, and both the rat fetal and adult kidney, with appropriate use of preimmune and preabsorption controls (27). Furthermore, this AT2 receptor antibody has been used to demonstrate AT2 receptor immunostaining in the myocardium and coronary vessels of the neonatal and young rat heart (41). Preabsorption against the peptide antigen blocked any positive immunostaining in these studies, and on immunoblots the AT2 receptor protein was detected in neonatal cardiac myocytes, but not neonatal cardiac fibroblasts or rat aortic smooth muscle cells (41). After incubation with the AT2 receptor antibody, the slides were incubated for 30 min at room temperature in a humidified chamber with a biotinylated anti-rabbit IgG, diluted 1:50 in PBS as a secondary antibody. The slides were subsequently placed in 3% H2O2 for 10 min prior to incubation with streptavidin-horseradish peroxidase (HRP), diluted 1:50 in PBS for 30 min at room temperature in a humidified chamber. Finally, the slides were incubated with 50 µl of diaminobenzadine (BioGenex, San Ramon, CA) as substrate. The slides were counterstained with hematoxylin (Sigma), dehydrated, fixed with Permount (Fisher Scientific, Ottawa, ON) histological mounting medium, and viewed with a Zeiss Axiophot microscope. To exclude nonspecific binding, all experiments included a control in which the primary antibody was preincubated with a 20-fold excess of immunizing peptide for 1 h at 37°C. Slides were quantitatively examined by using the Image-Pro Plus 4.0 software program (Media Cybernetics, Silver Spring, MD). Each slide was analyzed for staining in the cortex, outer medulla, and inner medulla, with three separate fields viewed in each region. Observations were made with the viewer blinded to the origin of the slide.
Western blot analysis of the AT1 and AT2
receptor.
Proteins from glomeruli of C, D, and D+I rats were isolated as
described (27). Briefly, glomeruli were homogenized in a buffer containing (in mM) 20 Tris · HCl, 100 NaCl, 2 phenylmethylsulfonyl fluoride, 2 EDTA, 2 EGTA, and 10 sodium
orthovanadate, as well as 10% glycerol, 10 µg/l leupeptin, and 10 µg/l aprotinin. The homogenate was then centrifuged at 30,000 g for 30 min at 4°C. The resulting pellet was resuspended
in the homogenization buffer, supplemented with 1% Nonidet P40, and
was stirred for 2 h at 4°C. The lysate was then centrifuged at
30,000 g for 30 min at 4°C. The supernatants were removed
and kept at 80°C until used for further analysis.
Statistical analysis. Results are expressed as means ± SE. Data were analyzed by one-way ANOVA followed by Bonferonni correction for all pairwise comparisons, or, in the case of the Western blots, by the Mann-Whitney rank-sum test for nonparametric data. A value of P < 0.05 was deemed significant.
![]() |
RESULTS |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Whole animal data.
Table 1 illustrates whole animal data for
C, D, and D+I rats. Diabetic rats were markedly hyperglycemic and
demonstrated glucosuria, similar to previous studies utilizing this
model (7, 11). In D+I rats, blood glucose values
decreased, and were, indeed, significantly lower than control values
(D+I: 4.9 ± 0.4 mM vs. C: 9.6 ± 0.2 mM; P < 0.05; n = 12). There were significant decreases in
body weight, and increases in kidney weights, in the diabetic group.
Insulin treatment completely reversed the decreases in body weight, as
well the increases in kidney weight. Serum creatinine levels did not
change in any of the groups. In the diabetic group, plasma renin
activity (PRA) was slightly suppressed, although this did not achieve
statistical significance compared with the control group.
|
Competitive RT-PCR.
Renin, angiotensinogen, and ACE mRNAs were quantitated from glomeruli
by competitive RT-PCR. As shown in Fig.
2, there was an increase in renin mRNA
levels in the glomeruli of D rats compared with C rats, although this
did not reach statistical significance (C: 2,497.5 ± 405.0 fg
mRNA/62.5 ng RNA vs. D: 3,155.5 ± 417.3 fg mRNA/62.5 ng RNA;
P = NS; n = 6). Moreover, there was no
significant difference in glomerular renin mRNA between C and D+I rats
(D+I: 2,490.0 ± 645.8 fg mRNA/62.5 ng RNA).
|
|
Glomerular AT1 receptor mRNA and protein expression.
By Northern analysis, AT1 receptor mRNA was readily
detected in glomeruli from C, D, and D+I rats, as a band at ~2.3 kb.
Early diabetes had no significant effect on glomerular AT1
receptor mRNA expression (Fig. 4).
Western blot analysis was performed on glomerular cell lysates for the
AT1 receptor. A band of the predicted molecular mass of the
nonglycosylated form of the AT1 receptor (41 kDa)
(13) was observed on all Western blots, with an additional
band at ~53 kDa, likely representing the glycosylated form of the
receptor (21). There was no significant change in expression of the 53-kDa glycosylated form of the AT1
receptor in glomeruli from diabetic rats [5.4 ± 2.8% increase;
P = not significant (NS) vs. C; n = 7], although expression of the 41-kDa nonglycosylated receptor protein
was significantly increased (Fig. 5: D:
354.4 ± 127.4% of C; P < 0.005 vs. C;
n = 7), with only partial reversal to C values in the
D+I group (D+I: 212.3 ± 41.1% of C; P < 0.05 vs. C; n = 7).
|
|
Western blot analysis of glomerular AT2 receptors.
By using RT-PCR, mRNA for AT2 receptors was inconsistently
detected in isolated glomeruli (4/7 preparations, data not shown), consistent with other studies indicating a low abundance of this message in adult kidney (30). By Western blot analysis,
however, a single band of the predicted molecular mass for the
AT2 receptor (44 kDa) was readily detected in cell lysates
from glomeruli, with no other bands observed (27). A
significant decrease in AT2 receptor protein expression was
observed in glomerular cell lysates (Fig.
6: D: 47.0 ± 6.5% of C;
P < 0.001 vs. C; n = 6). The decrease
in AT2 receptor expression was only partially reversed in
the insulin-implanted diabetic group (D+I: 66.8 ± 8.4% of C; P < 0.005 vs. C; n = 6).
|
AT2 receptor immunohistochemistry.
In the cortex of C rats, AT2-receptor immunostaining was
predominantly localized to glomerular endothelial cells, although diffuse staining of cortical tubular segments was also evident (Fig.
7A). In D rats, glomerular
staining for the AT2 receptor was markedly decreased (Fig.
7B), an effect consistently observed in all sections
(n = 8). Kidney sections from D+I rat sections displayed significant AT2 receptor staining within
glomeruli in all sections (n = 8; Fig. 7C)
to levels not significantly different from control. As a negative
control, no glomerular staining was observed in rat kidney sections in
which the primary antibody was preincubated with a 20-fold excess of
immunizing peptide, demonstrating the specificity of the
AT2 receptor antibody (Fig. 7D).
|
|
|
|
![]() |
DISCUSSION |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
In the present study, early diabetes had no significant effect on glomerular mRNA expression for renin, angiotensinogen, ACE, or AT1 receptors. The major findings are that diabetes was associated with a significant decrease in glomerular AT2 receptor expression, and, indeed, AT2 receptors were decreased throughout the diabetic kidney, as shown by immunohistochemistry. This effect was partially reversed by insulin implants. Our data suggest, therefore, that even if the intraglomerular RAS is not activated in early diabetes, the relative expression of AT1 and AT2 receptors may be important in defining the effects of ANG II in progressive diabetic nephropathy.
In these studies, we used the well-established STZ rat model of diabetes. In all rats, the characteristic increases in plasma glucose, decreases in body weight, and renal hypertrophy were observed after 2 wk (3, 11). In rats with insulin implants, there was a reversal of the effect on body weight, and renal hypertrophy, and plasma glucose levels were decreased compared with C rats, perhaps because of excess insulin administration. Moreover, and importantly, there was no significant change in PRA in the diabetic rats, which is consistent with other reports at this early time point of experimental diabetes (3, 11).
The glomerulus contains all components of the RAS necessary to produce ANG II (8, 19, 46). Indeed, Atiyeh et al. (5) showed that isolated rat glomeruli were capable of ANG II generation, an effect blocked by ACE inhibition in a concentration-dependent fashion. Tank et al. (36) have shown that glomerular renin mRNA is regulated by dietary salt ingestion, an effect reproduced in the present studies. However, no studies have focused on the effects of diabetes on RAS expression in the glomerulus, or on glomerular ANG II production. Accordingly, to determine whether early diabetes altered glomerular mRNA expression for components of the RAS, we used a quantitative competitive RT-PCR method. Preliminary experiments determined that the RT-PCR assay could detect changes in mRNA expression exceeding 20%. Our data suggest that glomerular mRNA expression for RAS components is not significantly altered in early diabetes. Alternatively, mRNA levels may be increased to levels below the detectable range for our assay. Indeed, renin mRNA levels were increased, although this did not reach statistical significance. Similarly, we have observed increased expression of proximal tubule renin mRNA after 2 wk in STZ diabetes (51), and increased whole kidney renin mRNA expression has been reported (3). It is also possible that glomerular ANG II levels could be increased in diabetes, independent of mRNA regulation, reflecting changes in mRNA translational efficiency or protein stability. Thus although we did not observe an effect on glomerular ACE mRNA, Anderson et al. (3) have demonstrated an increase in glomerular ACE protein in early STZ diabetes in rats.
Another means of regulation of the intrarenal RAS is at the level of expression of ANG II, AT1, and AT2 receptors in target tissue. In the kidney, AT1 receptors are present in abundance in vascular smooth muscle cells, endothelium, glomerular mesangial cells, podocytes, tubular cells along the entire nephron, and medullary interstitial cells (13, 31, 50). In contrast, AT2 receptor expression predominates in the fetal kidney and appears to diminish after birth (30). By using a polyclonal anti-AT2 receptor antibody (identical to the one used in the present studies) Ozono et al. (27) demonstrated AT2 receptor immunoreactivity in glomeruli, tubules, and interstitial cells from adult rats, with increased expression with sodium depletion. In contrast, immunohistochemical studies by Miyata et al. (24), by using a different antibody, revealed AT2 receptor expression in all nephron segments in adult rat kidney, except in the medullary thick ascending limb and the glomerulus. Our results indicate that AT2 receptors are expressed in the adult rat glomerulus, by both Western analysis and immunohistochemistry. Staining was observed in glomerular endothelial cells (Fig. 6), consistent with functional studies demonstrating effects of AT2 receptor activation in cultured glomerular endothelial cells (45). In addition, we observed AT2 receptor expression in cortical and outer medullary tubular segments, medullary interstitial cells, and IMCD cells. The reasons for differences in expression pattern compared with the study of Miyata et al. (24) are unclear but could be due to different antibody specificities or tissue preparations. Taken together, however, the data indicate that AT2 receptors are expressed in the adult kidney, and, furthermore, we demonstrated that preincubation of antibody with the immunizing antigen completely eliminated staining, indicating specificity of the antibody. The AT2 antibody used in the present studies has also been previously validated by its recognition of the AT2 receptor in stably transfected COS-7 cells and by its ability to detect AT2 receptors in a variety of adult and fetal tissues in the rat (27, 41).
Early diabetes caused a significant decrease in AT2 receptor protein expression, by both Western analysis and immunohistochemistry. In contrast, we observed no change in glomerular AT1 receptor mRNA, an unexpected increase in expression of the nonglycosylated 41-kDa AT1 receptor protein (an effect partly reversed by insulin), and no change in expression of the 53-kDa glycosylated receptor. It is noteworthy that decreased glomerular ANG II receptor density has been reported in early diabetes, with a persistent reduction in intrarenal AT1 receptors by radioligand binding up to 12 wk after STZ diabetes (7, 9, 16, 43). A decrease in AT1 receptor mRNA has also been demonstrated in whole kidney biopsy samples from humans with type II diabetes (40). In proximal tubule, AT1 receptor mRNA and protein are both decreased in early STZ diabetes (11, 51). The functional significance of alterations in the relative amounts of glycosylated and nonglycosylated AT1 receptors within diabetic glomeruli is unclear, although we speculate that this could alter AT1 receptor binding properties by affecting the numbers of receptors inserted into the plasma membrane.
In contrast to effects on AT1 receptor expression, we observed a marked decrease in glomerular, and, indeed, intrarenal AT2 receptor expression in early diabetes. The possible consequences of diminished AT2 receptor expression merit discussion. In a series of studies, Siragy and Carey (34) have demonstrated that AT2 receptors are linked to stimulation of intrarenal nitric oxide formation, increases in interstitial cGMP levels (33), and vasodilatation in a kidney-wrap model of hypertension (32). Activation of AT2 receptors also appears to counteract the hypertensive effects of AT1 receptor activation, as demonstrated in the AT2 receptor-knockout mouse (14). Given the present state of knowledge, it is difficult to predict the effects of decreased glomerular AT2 receptor expression on glomerular function in diabetes. However, decreased total intrarenal expression of AT2 receptors might promote enhanced tubular sodium reabsorption and increased blood pressure and could also lead to a decrease in AT2-mediated inhibition of cell growth (22, 26). This could result in an amplification of AT1-mediated effects on vasoconstriction or cell hypertrophy, contributing to increased nephron injury. In this regard, studies by Miller (23) in humans with early type I diabetes have demonstrated enhanced sensitivity of renal hemodynamic responses to AT1 receptor antagonism, suggesting a relative augmentation of intrarenal AT1-mediated receptor activity.
In our studies, insulin-implant therapy did not completely reverse the effects of STZ diabetes on AT1 and AT2 receptor expression in all regions. A number of possibilities for this must be considered. First, hyperglycemia may be only one of a number of factors that affect receptor expression in diabetes. Second, it is possible that even though glucose levels were corrected in the insulin implant group (and were indeed lower than controls), transient elevations of glucose may have occurred that could alter receptor expression. Finally, the direct effects of STZ on receptor expression must be considered, because this agent has been described to cause direct tubular nephrotoxicity (4). Notwithstanding these possibilities, the partial reversal with insulin indicates that high glucose at least partly exerts a regulatory effect on AT1 and AT2 receptor expression.
In summary, we have shown that early diabetes exerts no significant effect on mRNA expression of glomerular RAS components but significantly decreases AT2 receptors in glomeruli and other regions of the kidney. The results suggest that altered AT2 receptor expression in the kidney may be an important determinant of the rate of progression of diabetic nephropathy.
![]() |
ACKNOWLEDGEMENTS |
---|
The authors thank Dr. Susan Robertson (Dept. of Pathology, University of Ottawa) for assistance with histological examination of kidney AT2 receptors. Portions of this work were presented at the American Society of Nephrology meeting in Miami, FL, in November 1999 and have appeared in abstract form (J Am Soc Nephrol, 10: 691A, 1999).
![]() |
FOOTNOTES |
---|
This study was supported by a Diabetic Nephropathy Network Grant 996010 from the Medical Research Council of Canada /Juvenile Diabetes Foundation (K. D. Burns) and a grant from the Kidney Foundation of Canada (K. D. Burns.).
Address for reprint requests and other correspondence: K. D. Burns, Div. of Nephrology, Univ. of Ottawa and The Ottawa Hospital, 501 Smyth Rd., Rm. LM-18, Ottawa, Ontario, Canada K1H 8L6 (E-mail: kburns{at}ottawahospital.on.ca).
The costs of publication of this article were defrayed in part by the payment of page charges. The article must therefore be hereby marked "advertisement" in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.
Received 28 March 2000; accepted in final form 10 October 2000.
![]() |
REFERENCES |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
1.
Ahmad, J,
Siddiqui MA,
and
Ahmad H.
Effective postponement of diabetic nephropathy with enalapril in normotensive type 2 diabetic patients with microalbuminuria.
Diabetes Care
20:
1576-1581,
1997[Abstract].
2.
Akai, Y,
Homma T,
Burns KD,
Yasuda T,
Badr KF,
and
Harris RC.
Mechanical stretch/relaxation of cultured rat mesangial cells induces protooncogenes and cyclooxygenase.
Am J Physiol Cell Physiol
267:
C482-C490,
1994
3.
Anderson, S,
Jung FF,
and
Ingelfinger JR.
Renal renin-angiotensin system in diabetes: functional, immunohistochemical, and molecular biological correlations.
Am J Physiol Renal Fluid Electrolyte Physiol
265:
F477-F486,
1993
4.
Arison, RN,
Ciaccio EI,
Glitzer MS,
Cassaro JA,
and
Pruss MP.
Light and electron microscopy of lesions in rats rendered diabetic with streptozotocin.
Diabetes
16:
51-56,
1967[ISI][Medline].
5.
Atiyeh, BA,
Arant BS, Jr,
Henrich WL,
and
Seikaly MG.
In vitro production of angiotensin II by isolated glomeruli.
Am J Physiol Renal Fluid Electrolyte Physiol
268:
F266-F272,
1995
6.
Badr, KF,
DeBoer DK,
Takahashi K,
Harris RC,
Fogo A,
and
Jacobson HR.
Glomerular responses to platelet-activating factor in the rat: role of thromboxane A2.
Am J Physiol Renal Fluid Electrolyte Physiol
256:
F35-F43,
1989
7.
Ballermann, BJ,
Skorecki KL,
and
Brenner BM.
Reduced glomerular angiotensin II receptor density in early untreated diabetes mellitus in the rat.
Am J Physiol Renal Fluid Electrolyte Physiol
247:
F110-F116,
1984[ISI][Medline].
8.
Becker, BN,
Yasuda T,
Kondo S,
Vaikunth S,
Homma T,
and
Harris RC.
Mechanical stretch/relaxation stimulates a cellular renin-angiotensin system in cultured rat mesangial cells.
Exp Nephrol
6:
57-66,
1998[ISI][Medline].
9.
Brown, L,
Wall D,
Marchant C,
and
Sernia C.
Tissue-specific changes in angiotensin II receptors in streptozotocin-diabetic rats.
J Endocrinol
154:
355-362,
1997[Abstract].
10.
Burns, KD,
and
Smith IB.
Potassium depletion stimulates mRNA expression of proximal tubule AT1 angiotensin II receptors.
Nephron
78:
73-81,
1998[ISI][Medline].
11.
Cheng, HF,
Burns KD,
and
Harris RC.
Reduced proximal tubule angiotensin II receptor expression in streptozotocin-induced diabetes mellitus.
Kidney Int
46:
1603-1610,
1994[ISI][Medline].
12.
Dostal, DE,
Rothblum KN,
and
Baker KM.
An improved method for absolute quantification of mRNA using multiplex polymerase chain reaction: determination of renin and angiotensinogen mRNA levels in various tissues.
Anal Biochem
223:
239-250,
1994[ISI][Medline].
13.
Harrison-Bernard, LM,
Navar LG,
Ho MM,
Vinson GP,
and
el-Dahr SS.
Immunohistochemical localization of ANG II AT1 receptor in adult rat kidney using a monoclonal antibody.
Am J Physiol Renal Physiol
273:
F170-F177,
1997
14.
Ichiki, T,
Labosky PA,
Shiota C,
Okuyama S,
Imagawa Y,
Fogo A,
Niimura F,
Ichikawa I,
Hogan BL,
and
Inagami T.
Effects on blood pressure and exploratory behaviour of mice lacking angiotensin II type-2 receptor.
Nature
377:
748-750,
1995[ISI][Medline].
15.
Kagami, S,
Border WA,
Miller DE,
and
Noble NA.
Angiotensin II stimulates extracellular matrix protein synthesis through induction of transforming growth factor-beta expression in rat glomerular mesangial cells.
J Clin Invest
93:
2431-2437,
1994[ISI][Medline].
16.
Kalinyak, JE,
Sechi LA,
Griffin CA,
Don BR,
Tavangar K,
Kraemer FB,
Hoffman AR,
and
Schambelan M.
The renin-angiotensin system in streptozotocin-induced diabetes mellitus in the rat.
J Am Soc Nephrol
4:
1337-1345,
1993[Abstract].
17.
Koike, G,
Krieger JE,
Jacob HJ,
Mukoyama M,
Pratt RE,
and
Dzau VJ.
Angiotensin converting enzyme and genetic hypertension: cloning of rat cDNAs and characterization of the enzyme.
Biochem Biophys Res Commun
198:
380-386,
1994[ISI][Medline].
18.
Kontogiannis, J,
and
Burns KD.
Role of AT1 angiotensin II receptors in renal ischemic injury.
Am J Physiol Renal Physiol
274:
F79-F90,
1998
19.
Lai, KN,
Leung JC,
Lai KB,
To WY,
Yeung VT,
and
Lai FM.
Gene expression of the renin-angiotensin system in human kidney.
J Hypertens
16:
91-102,
1998[ISI][Medline].
20.
Lewis, EJ,
Hunsicker LG,
Bain RP,
and
Rohde RD.
The effect of angiotensin-converting-enzyme inhibition on diabetic nephropathy. The Collaborative Study Group
N Engl J Med
329:
1456-1462,
1993
21.
Li, X,
Shams M,
Zhu J,
Khalig A,
Wilkes M,
Whittle M,
Barnes N,
and
Ahmed A.
Cellular localization of AT1 receptor mRNA and protein in normal placenta and its reduced expression in intrauterine growth restriction.
J Clin Invest
101:
442-454,
1998
22.
Maric, C,
Aldred GP,
Harris PJ,
and
Alcorn D.
Angiotensin II inhibits growth of cultured embryonic renomedullary interstitial cells through the AT2 receptor.
Kidney Int
53:
92-99,
1998[ISI][Medline].
23.
Miller, JA.
Impact of hyperglycemia on the renin angiotensin system in early human type 1 diabetes mellitus.
J Am Soc Nephrol
10:
1778-1785,
1999
24.
Miyata, N,
Park F,
Li XF,
and
Cowley AW, Jr.
Distribution of angiotensin AT1 and AT2 receptor subtypes in the rat kidney.
Am J Physiol Renal Physiol
277:
F437-F446,
1999
25.
Mizuiri, S,
Yoshikawa H,
Tanegashima M,
Miyagi M,
Kobayashi M,
Sakai K,
Hayashi I,
Aikawa A,
Ohara T,
and
Hasegawa A.
Renal ACE immunohistochemical localization in NIDDM patients with nephropathy.
Am J Kidney Dis
31:
301-307,
1998[ISI][Medline].
26.
Nakajima, M,
Hutchinson HG,
Fujinaga M,
Hayashida W,
Morishita R,
Zhang L,
Horiuchi M,
Pratt RE,
and
Dzau VJ.
The angiotensin II type 2 (AT2) receptor antagonizes the growth effects of the AT1 receptor: gain-of-function study using gene transfer.
Proc Natl Acad Sci USA
92:
10663-10667,
1995[Abstract].
27.
Ozono, R,
Wang ZQ,
Moore AF,
Inagami T,
Siragy HM,
and
Carey RM.
Expression of the subtype 2 angiotensin (AT2) receptor protein in rat kidney.
Hypertension
30:
1238-1246,
1997
28.
Ravid, M,
Lang R,
Rachmani R,
and
Lishner M.
Long-term renoprotective effect of angiotensin-converting enzyme inhibition in non-insulin-dependent diabetes mellitus.
Arch Intern Med
156:
286-289,
1996[Abstract].
29.
Remuzzi, A,
Perico N,
Amuchastegui CS,
Malanchini B,
Mazerska M,
Battaglia C,
Bertani T,
and
Remuzzi G.
Short- and long-term effect of angiotensin II receptor blockade in rats with experimental diabetes.
J Am Soc Nephrol
4:
40-49,
1993[Abstract].
30.
Shanmugam, S,
Llorens-Cortes C,
Clauser E,
Corvol P,
and
Gasc JM.
Expression of angiotensin II AT2 receptor mRNA during development of rat kidney and adrenal gland.
Am J Physiol Renal Fluid Electrolyte Physiol
268:
F922-F930,
1995
31.
Sharma, M,
Sharma R,
Greene AS,
McCarthy ET,
and
Savin VJ.
Documentation of angiotensin II receptors in glomerular epithelial cells.
Am J Physiol Renal Physiol
274:
F623-F637,
1998
32.
Siragy, HM,
and
Carey RM.
Protective role of the angiotensin AT2 receptor in a renal wrap hypertension model.
Hypertension
33:
1237-1242,
1999
33.
Siragy, HM,
and
Carey RM.
The subtype-2 (AT2) angiotensin receptor regulates renal cyclic guanosine 3', 5'-monophosphate and AT1 receptor-mediated prostaglandin E2 production in conscious rats.
J Clin Invest
97:
1978-1982,
1996
34.
Siragy, HM,
and
Carey RM.
The subtype 2 (AT2) angiotensin receptor mediates renal production of nitric oxide in conscious rats.
J Clin Invest
100:
264-269,
1997
35.
Siragy, HM,
Inagami T,
Ichiki T,
and
Carey RM.
Sustained hypersensitivity to angiotensin II and its mechanism in mice lacking the subtype-2 (AT2) angiotensin receptor.
Proc Natl Acad Sci USA
96:
6506-6510,
1999
36.
Tank, JE,
Henrich WL,
and
Moe OW.
Regulation of glomerular and proximal tubule renin mRNA by chronic changes in dietary NaCl.
Am J Physiol Renal Physiol
273:
F892-F898,
1997
37.
Viberti, G,
Mogensen CE,
Groop LC,
and
Pauls JF.
Effect of captopril on progression to clinical proteinuria in patients with insulin-dependent diabetes mellitus and microalbuminuria.
JAMA
271:
275-279,
1994[Abstract].
38.
Villa, E,
Rabano A,
Cazes M,
Cloarec A,
Ruilope LM,
and
Garcia-Robles R.
Effects of UP269-6, a new angiotensin II receptor antagonist, and captopril on the progression of rat diabetic nephropathy.
Am J Hypertens
10:
275-281,
1997[ISI][Medline].
39.
Vinay, P,
Gougoux A,
and
Lemieux G.
Isolation of a pure suspension of rat proximal tubules.
Am J Physiol Renal Fluid Electrolyte Physiol
241:
F403-F411,
1981
40.
Wagner, J,
Gehlen F,
Ciechanowicz A,
and
Ritz E.
Angiotensin II receptor type 1 gene expression in human glomerulonephritis and diabetes mellitus.
J Am Soc Nephrol
10:
545-551,
1999
41.
Wang, ZQ,
Moore AF,
Ozono R,
Siragy HM,
and
Carey RM.
Immunolocalization of subtype angiotensin II (AT2) receptor protein in rat heart.
Hypertension
32:
78-83,
1998
42.
Wang, TT,
Wu XH,
Zhang SL,
and
Chan JS.
Effect of glucose on the expression of the angiotensinogen gene in opossum kidney cells.
Kidney Int
53:
312-319,
1998[ISI][Medline].
43.
Wilkes, BM.
Reduced glomerular angiotensin II receptor density in diabetes mellitus in the rat: time course and mechanism.
Endocrinology
120:
1291-1298,
1987[Abstract].
44.
Wolf, G,
Ziyadeh FN,
and
Stahl RA.
Angiotensin II stimulates expression of transforming growth factor beta receptor type II in cultured mouse proximal tubular cells.
J Mol Med
77:
556-564,
1999[ISI][Medline].
45.
Wolf, G,
Ziyadeh FN,
Thaiss F,
Tomaszewski J,
Caron RJ,
Wenzel U,
Zahner G,
Helmchen U,
and
Stahl RA.
Angiotensin II stimulates expression of the chemokine RANTES in rat glomerular endothelial cells. Role of the angiotensin type 2 receptor.
J Clin Invest
100:
1047-1058,
1997
46.
Wolf, G,
Ziyadeh FN,
Zahner G,
and
Stahl RA.
Angiotensin II is mitogenic for cultured rat glomerular endothelial cells.
Hypertension
27:
897-905,
1996
47.
Yamada, T,
Horiuchi M,
and
Dzau VJ.
Angiotensin II type 2 receptor mediates programmed cell death.
Proc Natl Acad Sci USA
93:
156-160,
1996
48.
Yuan, BH,
Robinette JB,
and
Conger JD.
Effect of angiotensin II and norepinephrine on isolated rat afferent and efferent arterioles.
Am J Physiol Renal Fluid Electrolyte Physiol
258:
F741-F750,
1990
49.
Zatz, R,
Dunn BR,
Meyer TW,
Anderson S,
Rennke HG,
and
Brenner BM.
Prevention of diabetic glomerulopathy by pharmacological amelioration of glomerular capillary hypertension.
J Clin Invest
77:
1925-1930,
1986[ISI][Medline].
50.
Zhuo, J,
Maric C,
Harris PJ,
Alcorn D,
and
Mendelsohn FA.
Localization and functional properties of angiotensin II AT1 receptors in the kidney: focus on renomedullary interstitial cells.
Hypertens Res
20:
233-250,
1997[Medline].
51.
Zimpelmann J, Kumar D, Levine DZ, Wehbi G, Imig JD, Navar LG, and Burns
KD. Early diabetes mellitus stimulates proximal tubule renin mRNA
expression in the rat. Kidney Int (in
press).