ArticlePDF Available

Intracellular Angiotensin II Production in Diabetic Rats Is Correlated With Cardiomyocyte Apoptosis, Oxidative Stress, and Cardiac Fibrosis

Authors:

Abstract and Figures

Many of the effects of angiotensin (Ang) II are mediated through specific plasma membrane receptors. However, Ang II also elicits biological effects from the interior of the cell (intracrine), some of which are not inhibited by Ang receptor blockers (ARBs). Recent in vitro studies have identified high glucose as a potent stimulus for the intracellular synthesis of Ang II, the production of which is mainly chymase dependent. In the present study, we determined whether hyperglycemia activates the cardiac intracellular renin-Ang system (RAS) in vivo and whether ARBs, ACE, or renin inhibitors block synthesis and effects of intracellular Ang II (iAng II). Diabetes was induced in adult male rats by streptozotocin. Diabetic rats were treated with insulin, candesartan (ARB), benazepril (ACE inhibitor), or aliskiren (renin inhibitor). One week of diabetes significantly increased iAng II levels in cardiac myocytes, which were not normalized by candesartan, suggesting that Ang II was synthesized intracellularly, not internalized through AT(1) receptor. Increased intracellular levels of Ang II, angiotensinogen, and renin were observed by confocal microscopy. iAng II synthesis was blocked by aliskiren but not by benazepril. Diabetes-induced superoxide production and cardiac fibrosis were partially inhibited by candesartan and benazepril, whereas aliskiren produced complete inhibition. Myocyte apoptosis was partially inhibited by all three agents. Diabetes activates the cardiac intracellular RAS, which increases oxidative stress and cardiac fibrosis. Renin inhibition has a more pronounced effect than ARBs and ACE inhibitors on these diabetes complications and may be clinically more efficacious.
Schematic representation of the relationship between hyperglycemia, iAng II, and pathological effects. In hyperglycemia, there is an increase in glucose oxidation through the tricarboxylic acid cycle in mitochondria, which results in enhanced generation of reactive oxygen species. Overproduction of superoxide inhibits glyceraldehyde-3-phosphate dehydrogenase activity, resulting in an accumulation of upstream metabolites of the glycolytic pathway. Increased levels of glyceraldehyde-3-phosphate (GAD-3P) cause activation of PKC isoforms through diacylglycerol (DAG) production and synthesis of advanced glycation end products (AGEs). There is increased shuttling of glucose through the hexosamine biosynthesis pathway, resulting in the modification of transcription factors through o-glycosylation. All of these products of hyperglycemia, i.e., oxidative stress, AGEs, PKC, and o-glycosylation of transcription factors, activate expression of RAS components. Cardiac myocytes synthesize and retain Ang II intracellularly in hyperglycemia, whereas cardiac fibroblasts increase both intra-and extracellular Ang II. iAng II could directly increase oxidative stress and cellular apoptosis through unidentified mechanisms and/or could enhance expression of RAS components through a positive feedback mechanism, resulting in enhanced extracellular Ang II levels as well, particularly via cardiac fibroblasts. Extracellular Ang II in turn causes oxidative stress, cardiac myocyte apoptosis, and cardiac fibrosis through the AT 1 receptor. Interrupting this cycle by blocking Ang II synthesis provides protection from hyperglycemia-induced pathological events. ACE inhibitors or ARBs would block only the extracellular synthesis or actions, respectively, of Ang II; whereas a renin inhibitor would block both intra-and extracellular Ang II synthesis, the latter providing an explanation for the more pronounced effects of aliskiren observed in this study.
… 
Content may be subject to copyright.
Intracellular Angiotensin II Production in Diabetic Rats
Is Correlated With Cardiomyocyte Apoptosis, Oxidative
Stress, and Cardiac Fibrosis
Vivek P. Singh,
1,2
Bao Le,
2
Renu Khode,
3
Kenneth M. Baker,
1,2
and Rajesh Kumar
1,2
OBJECTIVE—Many of the effects of angiotensin (Ang) II are
mediated through specific plasma membrane receptors. How-
ever, Ang II also elicits biological effects from the interior of the
cell (intracrine), some of which are not inhibited by Ang receptor
blockers (ARBs). Recent in vitro studies have identified high
glucose as a potent stimulus for the intracellular synthesis of Ang
II, the production of which is mainly chymase dependent. In the
present study, we determined whether hyperglycemia activates
the cardiac intracellular renin-Ang system (RAS) in vivo and
whether ARBs, ACE, or renin inhibitors block synthesis and
effects of intracellular Ang II (iAng II).
RESEARCH DESIGN AND METHODS—Diabetes was in-
duced in adult male rats by streptozotocin. Diabetic rats were
treated with insulin, candesartan (ARB), benazepril (ACE inhib-
itor), or aliskiren (renin inhibitor).
RESULTS—One week of diabetes significantly increased iAng II
levels in cardiac myocytes, which were not normalized by
candesartan, suggesting that Ang II was synthesized intracellu-
larly, not internalized through AT
1
receptor. Increased intracel-
lular levels of Ang II, angiotensinogen, and renin were observed
by confocal microscopy. iAng II synthesis was blocked by
aliskiren but not by benazepril. Diabetes-induced superoxide
production and cardiac fibrosis were partially inhibited by can-
desartan and benazepril, whereas aliskiren produced complete
inhibition. Myocyte apoptosis was partially inhibited by all three
agents.
CONCLUSIONS—Diabetes activates the cardiac intracellular
RAS, which increases oxidative stress and cardiac fibrosis. Renin
inhibition has a more pronounced effect than ARBs and ACE
inhibitors on these diabetes complications and may be clinically
more efficacious. Diabetes 57:3297–3306, 2008
Involvement of the renin-angiotensin (Ang) system
(RAS) in human pathophysiology has expanded to
include several diseases beyond a traditional role in
saltwater homeostasis (1). In diabetes, there is sig-
nificant overactivity of the RAS, which is reversed by
treatment with RAS inhibitors, thus decreasing diabetes
complications (2). Activation of the RAS in diabetes in-
cludes activation of new components, such as the pro(re-
nin) receptor (3), and Ang II–independent effects,
mediated through interaction of pro(renin), with the pro-
(renin) receptor (4). Although circulating renin and Ang II
levels are reduced in diabetes, prorenin levels are en-
hanced severalfold (5,6). Prorenin may have dual effects,
providing for generation of Ang I at tissue sites through
receptor-mediated nonproteolytic activation and directly
through activation of receptor-mediated signaling path-
ways (4,7,8). Ang II–independent RAS actions suggest that
efficacy of RAS inhibitors, Ang receptor blockers (ARBs),
and ACE inhibitors would have limitations in hyperglyce-
mic conditions. Recent meta-analyses of clinical trials
have suggested that currently used RAS blockers may not
provide additional benefits in diabetic compared with
nondiabetic patients (9,10).
We recently reported a novel aspect of the RAS, the
intracellular RAS, having identified an intracellular or
intracrine system (11,12). In cardiac myocytes and fibro-
blasts, we demonstrated the presence of RAS components
and synthesis of Ang II intracellularly (13,14). Hyperglyce-
mia selectively upregulates the intracellular system in
cardiac myocytes, vascular smooth muscle cells (VSMCs),
and renal mesangial cells, where Ang II synthesis is largely
catalyzed by chymase, not ACE (14 –18). We and others
have previously reported that intracellular Ang II (iAng II)
elicits biological effects, some of which are not blocked by
ARBs (19 –22). These observations further support the
speculation that currently available RAS inhibitors may
not provide the anticipated cardiovascular benefits in
diabetic conditions (23). In this study, we have examined
the activation of the cardiac intracellular RAS in a rat
model of diabetes. We also determined the role of iAng II
in diabetes-induced oxidative stress, cardiac myocyte apop-
tosis, and cardiac fibrosis and the efficacy of different RAS
blockers under hyperglycemic conditions.
RESEARCH DESIGN AND METHODS
All animal use was approved by the Institutional Animal Care and Use
Committee of the Texas A&M Health Science Center. The AT
1
receptor
blocker candesartan was obtained from AstraZeneca (Wilmington, DE); the
renin inhibitor aliskiren was from Novartis (Cambridge, MA); the ACE
inhibitor benazepril was from Sigma; and insulin (Humulin N) was from Eli
Lilly (Indianapolis, IN).
Induction of diabetes and treatment of animals. Diabetes was induced by
a single injection of streptozotocin (STZ, 65 mg/kg body wt i.p.) dissolved in
0.1 mol/l sodium citrate– buffered saline (pH 4.5), in adult male Sprague
Dawley rats (250 –300 g). Control animals received buffered saline alone.
Diabetes was confirmed by sustained blood glucose levels 15 mmol/l, as
determined 48 h after STZ injection and on alternate days thereafter. Diabetic
rats, in groups of nine animals, were treated with insulin (2–5 units s.c., twice
daily), candesartan (1 mg/kg i.p.), aliskiren (30 mg/kg orally), or benazepril (10
mg/kg orally) daily for 7 days beginning 48 h after STZ injection. Twenty-four
hours after the last treatment, animals were weighed and anesthetized using
From the
1
Division of Molecular Cardiology, Department of Medicine, Texas
A&M Health Science Center, College of Medicine, Scott & White, Central
Texas Veterans Health Care System, Temple, Texas; and the
2
Department of
Medicine, Scott & White, Temple, Texas; and
3
Department of Pathology,
Scott & White, Temple, Texas.
Corresponding author: Rajesh Kumar, kumar@medicine.tamhsc.edu.
Received 17 June 2008 and accepted 8 September 2008.
Published ahead of print at http://diabetes.diabetesjournals.org on 1 October
2008. DOI: 10.2337/db08-0805.
© 2008 by the American Diabetes Association. Readers may use this article as
long as the work is properly cited, the use is educational and not for profit,
and the work is not altered. See http://creativecommons.org/licenses/by
-nc-nd/3.0/ for details.
The costs of publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked “advertisement” in accordance
with 18 U.S.C. Section 1734 solely to indicate this fact.
ORIGINAL ARTICLE
DIABETES, VOL. 57, DECEMBER 2008 3297
ketamine/xylazine (50/5 mg/kg), and hearts were isolated and weighed before
perfusion, the latter using the Langendorff methodology.
Isolation of cardiac myocytes and measurement of iAng II. Hearts were
isolated and perfused with Krebs-Henseleit bicarbonate buffer, followed by
digestion with 0.1% (wt/vol) collagenase II. Myocytes were separated from
nonmyocytes by differential centrifugation at 25g. The purity of the myocyte
preparations using this procedure was 90%, as analyzed by fluorescence-
activated cell sorting, using anti-sarcomeric myosin (MF-20) and anti-sarco-
meric actin antibody. The pellet containing myocytes was processed for Ang
II extraction, as described previously (14). Briefly, cells were lysed in ice-cold
1 mol/l acetic acid containing a protease inhibitor cocktail (Sigma) by brief
sonication. The lysate was sedimented at 20,000gfor 10 min, and the
supernatant was dried in a vacufuge, followed by reconstitution in 1% acetic
acid. The samples were applied to a conditioned DSC-18 column (Supelco),
washed, and eluted with methanol. The eluted samples were dried and reconsti-
tuted in PBS for enzyme-linked immunosorbent assay (ELISA). For isolation of
Ang II from plasma, an equal volume of 2% acetic acid was added to plasma,
followed by filtration through Amicon Ultra-15 filters. The filtrate was applied to
DSC-18 columns, and Ang II was eluted as described for the cell lysates. Using the
above procedure, we have obtained 90% recovery of exogenously added Ang II.
Ang II was measured by quantitative, competitive ELISA, using a specific
anti–Ang II antibody (Peninsula Labs), which was previously validated by
high-performance liquid chromatography– chip/mass spectrometric analysis (14).
ELISA was performed on protein-A and anti–Ang II antibody– coated 96-well
dishes. Competitive binding of synthetic biotinylated Ang II, in the presence of the
extracted peptide, was detected with streptavidin– horseradish peroxidase con-
jugate. A standard curve, generated from binding of a constant amount of
biotinylated Ang II with increasing concentrations of nonbiotinylated synthetic
Ang II, was used to calculate the concentration of the peptide in the sample. The
concentration of Ang II in the cell lysates is expressed as femtomoles per
milligram protein and in plasma as femtomoles per milliliter.
Immunohistochemistry of RAS components. Hearts were frozen in OCT
compound (Tissue-Tek; Sakura Finetek) at 80°C for immunofluorescence
staining of Ang II, renin, and anti-angiotensinogen (AGT). Frozen tissue was
cut into 5-m sections, which were air-dried, fixed with 4% formaldehyde, and
permeabilized using 0.2% Triton X-100. Nonspecific binding was blocked by 5%
BSAfor1hatroom temperature. The sections were incubated with anti–Ang
II antibody (1:100; Peninsula Labs), anti-renin antibody (1:100; gift from Dr.
Tadashi Inagami [Vanderbilt University, Nashville, TN]), or AGT antibody
(1:500; Swant). The sections were costained for anti-sarcomeric actin and
laminin, where indicated. After washings, the sections were incubated with
respective secondary antibodies. Specificity of the staining was determined by
preadsorption of primary antibodies with the antigen or by using secondary
antibody alone. Images were acquired with a confocal fluorescence micro-
scope (Olympus Fluoview 300). Fluorescence intensities in tissue sections
were determined by digital microscopy software (Slide Book 4.2) after
subtracting background fluorescence.
Cardiac myocyte apoptosis. Apoptotic cardiac myocytes were detected in
paraffin-embedded heart sections using the terminal deoxynucleotide trans-
ferase-mediated dUTP nick-end labeling (TUNEL) assay and cleaved
caspase-3 staining. TUNEL assay was performed using an assay kit (Millipore,
Temecula, CA) per the manufacturer’s instructions. Cytoplasm and nuclei
from the myocytes were counterstained using anti-sarcomeric actin antibody
and DAPI, respectively. For cleaved caspase-3 staining, deparaffinized sec-
tions were subjected to antigen retrieval in 0.01 mol/l citrate buffer (pH 6.0) by
microwaving. After blocking with 5% BSA, the sections were incubated with
rabbit monoclonal anti– cleaved caspase-3 antibody (1:200; Cell Signaling
Technology, Danvers, MA) overnight at 4°C, followed by fluorescein isothio-
cyanate– conjugated goat anti-rabbit IgG (1:200; Molecular Probes). The
number of positively stained nuclei was counted from 20 fields per heart
(25,000 cells) and three hearts per treatment group.
Reactive oxygen species detection in the heart. Superoxide production in
the hearts was detected by dihydroethidium (DHE) staining (Sigma-Aldrich).
Frozen heart sections (20 m thick) were incubated with 10 mol/l DHE at
37°C for 45 min in a humidified chamber protected from light. Fluorescent
images obtained with an Olympus FV300 confocal microscope were analyzed
with Slide Book 4.2. The mean DHE fluorescence intensity of myocyte nuclei
was calculated by dividing the combined fluorescence value of the pixels by
the total number of pixels in 15 randomly selected fields observed with
identical laser and photomultiplier settings.
Cardiac fibrosis. Cardiac interstitial fibrosis was determined by Masson’s
trichrome staining on 5-m paraffin-embedded sections. The extent and
degree of fibrosis was subjectively graded on a scale of 0 –4. Grade 0 signified
no apparent collagen fiber proliferation except for small islets of fibrous tissue
around the capillaries, as well as an intercellular single layer of collagenous
tissue, as in normal myocardium. Focal and minimal fibrosis was graded as 1,
mild patchy fibrosis as grade 2, moderate diffuse fibrosis as grade 3, and the
most prominent fibrosis, covering a major area of the specimen, was classified
as 4. A minimum of three sections per heart with five fields per section and
three animals per experimental group were analyzed, and results are pre-
sented as an average grade.
Statistical analysis. Values are expressed as means SE. ANOVA with
Tukey’s post hoc test was used for statistical analysis. P0.05 was
considered statistically significant.
RESULTS
Hyperglycemia increases intracellular levels of Ang
II in cardiac myocytes from diabetic heart. Diabetes
was induced in adult male rats by STZ injection. One group
of diabetic animals was treated with insulin to confirm that
the observed effects in the experimental groups were
secondary to hyperglycemia. One week of diabetes signif-
icantly reduced body and heart weights, which were
normalized by insulin treatment but not by any of the RAS
inhibitors (Table 1). However, no significant effect on the
heart-to-body weight ratio (Table 1) or plasma Ang II
levels (Fig. 1B) was observed, which is consistent with
previous reports (24,25). Ang II levels in cardiac myocytes,
which were isolated after perfusion of the hearts and
enzymatic dispersion, represented Ang II present intracel-
lularly. To determine the source of iAng II, i.e., intracellu-
lar synthesis or AT
1
-mediated internalization, one group of
diabetic animals was treated with the AT
1
antagonist
candesartan to prevent receptor-mediated uptake. As
shown in Fig. 1A, cardiac myocytes from diabetic rat
hearts demonstrated a 9.4-fold elevation in the levels of
iAng II (183 13 fmol/mg protein) compared with cells
from control animals (19 4 fmol/mg protein). Normal-
ization of blood glucose levels by insulin in rats adminis-
tered STZ completely blocked the rise in iAng II levels,
indicating that the latter was a specific effect of hyper-
glycemia. Treatment of diabetic rats with candesartan
partially reduced iAng II levels, suggesting that the major
source of iAng II was intracellular synthesis, which is
consistent with our previous report in neonatal rat ven-
tricular myocytes (NRVMs) (14).
Intracellular synthesis of Ang II is not blocked by an
ACE inhibitor. We and others had previously reported
that several cell types (NRVMs, VSMCs, and renal mesan-
TABLE 1
Effect of diabetes on body and heart weight
Control
Diabetic
No
treatment Insulin Aliskiren Candesartan Benazepril
Body weight (g) 337 10 263 8.3* 307 4.7 248 8.5* 285 5.9* 267 6.9*
Heart weight (mg) 1,168 54 933 33* 1,074 27 867 37* 988 15* 901 16*
Heart weight/body weight (mg/g) 3.46 0.1 3.55 0.1 3.49 0.1 3.50 0.1 3.47 0.1 3.37 0.1
Data are means SE (n9) after 1 week of induction of diabetes. *P0.05 vs. control.
INTRACELLULAR Ang II IN DIABETES
3298 DIABETES, VOL. 57, DECEMBER 2008
gial cells) use alternative pathways to synthesize Ang II in
high-glucose culture conditions (14 –18). To determine the
mechanism of hyperglycemia-induced cardiac iAng II syn-
thesis in vivo, diabetic rats were treated with either a renin
inhibitor (aliskiren) or an ACE inhibitor (benazepril). As
shown in Fig. 1A, aliskiren completely normalized (33 7
fmol/mg protein) iAng II levels in diabetic rat cardiac
myocytes, whereas benazepril did not have any effect
(143 8 fmol/mg protein). These results indicated that the
observed increase in iAng II was not catalyzed by ACE,
which is consistent with intracellular synthesis of Ang II.
None of the RAS blocking drugs significantly altered
plasma Ang II levels, which were measured 24 h after the
last dose. The latter observation corroborated with the
reported 2- to 6-h period for reactive changes in plasma
levels of Ang II, which returned to baseline between 14
and 30 h after drug intake (26 –28). The lack of increase in
plasma Ang II levels in any of the treatment groups further
indicated that the increase in cardiac myocyte Ang II levels
was due to local synthesis.
Immunohistochemical localization of Ang II in rat
heart. To further confirm elevation of Ang II in cardiac
myocytes and intracellular localization, frozen heart sec-
tions were immunostained with anti–Ang II antibody and
visualized using confocal microscopy. Sections were coun-
terstained for laminin to mark cell boundaries (peripheral
green/yellow staining in merged images), anti-sarcomeric
actin to identify cardiac myocytes (red), and DAPI to
identify nuclei (blue). Significantly increased levels of Ang
II staining, which colocalized with anti-sarcomeric actin,
were observed in diabetic rat hearts (Fig. 1C–F). Quanti-
fication of fluorescence intensity revealed about a fivefold
FIG. 1. Ang II levels in cardiac myocytes and plasma. Ang II was measured by a competitive ELISA in cardiac myocytes (A) and plasma (B)of
control rats (Cont); diabetic rats (STZ); and diabetic rats treated with insulin (Ins), aliskiren (Alsk), candesartan (Cand), or benazepril (Bnz).
Values are expressed as means SE, n6. C–F: Intracellular localization of Ang II (yellow dots, indicated by white arrow), as determined by
confocal immunofluorescence microscopy, in heart sections from control rats (C), diabetic rats (D), diabetic rats treated with aliskiren (E), and
diabetic rats treated with candesartan (F). Myocyte profiles were identified by costaining with anti-sarcomeric actin (red) and laminin (yellow,
peripheral staining). The blue color indicates nuclear staining by DAPI. Magnification 1,200. G: Quantitative representation of Ang II
fluorescence intensity in heart sections (from five images per heart and three hearts per group). Values are expressed as means SE, n15.
*P<0.05 vs. control, †P<0.05 vs. diabetic rats without any treatment. (Please see http://dx.doi.org/10.2337/db08-0805 for a high-quality digital
representation of this figure.)
V.P. SINGH AND ASSOCIATES
DIABETES, VOL. 57, DECEMBER 2008 3299
increase in iAng II levels (Fig. 1G), consistent with the Ang
II measurement by ELISA (Fig. 1A). Immunohistochemis-
try also confirmed that candesartan partially reduced iAng
II levels, benazepril did not have any effect, and aliskiren
completely prevented the increase in iAng II in diabetic
hearts (Fig. 1G).
Intracellular localization of AGT and renin in cardiac
myocytes. Intracellular synthesis of Ang II would require
the presence of the precursor molecule AGT and process-
ing enzyme, renin, intracellularly. To demonstrate this
possibility, immunohistochemistry was performed on
heart sections using anti-AGT and anti-renin antibodies,
along with counterstaining for anti-sarcomeric actin, as
described previously (24). Elevated levels of AGT and
renin were apparent in cardiac myocytes of diabetic hearts
(green/yellow staining) compared with control, confirming
activation of the intracellular RAS in hyperglycemic con-
ditions (Fig. 2).
iAng II is correlated with hyperglycemia-induced
oxidative stress. Hyperglycemia is known to induce
myocardial oxidative stress, which may be related to
glucose metabolism or activation of cytokines and other
hormones. To determine whether there was a role of iAng
II, superoxide production was detected by DHE staining in
frozen heart sections of diabetic rats treated with different
RAS inhibitors. As shown in Fig. 3, diabetic hearts showed
enhanced superoxide production, which was prevented
in insulin-treated animals. Treatment of diabetic rats
with candesartan or benazepril significantly, but not
completely, reduced oxidative stress, whereas aliskiren
blocked completely. Our previous studies had indicated
that ARBs and ACE inhibitors were ineffective in block-
ing the intracellular RAS, unlike a renin inhibitor that
blocks both the intracellular and extracellular systems
(14,19,22). The observed partial efficacy of candesartan
and benazepril strongly suggested that iAng II contrib-
uted to hyperglycemia-induced oxidative stress in the
myocardium.
iAng II is correlated with hyperglycemia-induced
cardiac myocyte apoptosis. Cardiac myocyte apoptosis
was determined by TUNEL assay and activated caspase-3
immunostaining. Figure 4 shows an increased number of
apoptotic cells in the heart sections. Quantification of
apoptotic cells (Fig. 4Cand F) showed a five- to eightfold
increase in diabetic hearts compared with control by both
TUNEL assay and caspase-3 staining. Normalization of
blood glucose by insulin or blockade of the RAS with the
three different inhibitors significantly reduced the num-
ber of apoptotic cells but did not prevent apoptosis
completely. There was a significant difference between
aliskiren- and benazepril-treated animals, with aliskiren
being more protective.
iAng II is correlated with hyperglycemia-induced
cardiac fibrosis. Cardiac fibrosis is an important patho-
genic factor in diabetes-induced diastolic dysfunction.
Paraffin-embedded heart sections were stained with Mas-
son’s Trichrome, and the degree of blue staining was
evaluated on a scale of 0 4, as described in RESEARCH DESIGN
FIG. 2. Representative confocal immunofluorescence images of AGT and renin staining in hearts from control and diabetic rats. Pictures shown
are merged images of staining for anti-sarcomeric actin (red), laminin (peripheral red staining in top), AGT (top) or renin (bottom) (green and
yellow staining), and nuclei (blue). Magnification 900. (Please see http://dx.doi.org/10.2337/db08-0805 for a high-quality digital representation
of this figure.)
INTRACELLULAR Ang II IN DIABETES
3300 DIABETES, VOL. 57, DECEMBER 2008
AND METHODS. Even after only 1 week of diabetes, the overall
staining for fibrosis was enhanced in hearts from diabetic
rats (grade 1.5) compared with control animals (grade 0)
(Fig. 5). Insulin treatment completely prevented the in-
crease in fibrosis (grade 0.04). Candesartan and benazepril
reduced the degree of fibrosis (grade 0.43 and 0.88, respec-
FIG. 3. Measurement of oxidative stress in heart
sections by DHE staining. These are representative
images of DHE-stained heart sections from control
rats (A); diabetic rats (B); and diabetic rats treated
with insulin (C), aliskiren (D), candesartan (E), or
benazepril (F). Magnification 60. G: DHE fluores-
cence intensity was calculated from five images per
heart and three hearts per group. Values are ex-
pressed as means SE (n15). *P<0.05 vs.
control, †P<0.05 vs. diabetic rats without any
treatment. (Please see http://dx.doi.org/10.2337/
db08-0805 for a high-quality digital representation
of this figure.)
A B
DE
STZ
0
0.1
0.2
0.3
0.4
Cont Ins Alsk Cand Bnz
Activated Caspase- 3
Positive Cells (%)
F
*
*
*
*
*
*
0
0.1
0.2
0.3
0.4
Cont Ins Alsk Cand Bnz
TUNEL Positive Nuclei
(%)
*
*
*
*
STZ
C
FIG. 4. Detection of apoptosis in cardiac myocytes by TUNEL assay and cleaved caspase-3 staining. Aand B: TUNEL assay on heart sections from control (A)
and diabetic (B) rats. Dand E: Staining for cleaved caspase-3 in the hearts of control (D) and diabetic (E) rats. Cand F: Quantification of TUNEL
(C) and
cleaved caspase-3
(F) cells (25,000 cells were counted in each case). Values are expressed as means SE. *P<0.05 vs. control, †P<0.05 vs. diabetic rats
without any treatment. (Please see http://dx.doi.org/10.2337/db08-0805 for a high-quality digital representation of this figure.)
V.P. SINGH AND ASSOCIATES
DIABETES, VOL. 57, DECEMBER 2008 3301
tively), whereas aliskiren had a more pronounced reduc-
tion of fibrosis (grade 0.25) in diabetic rat hearts (Fig. 5).
DISCUSSION
In this study, we observe a dramatic activation of the
intracellular RAS, a novel aspect of the tissue RAS, in
diabetic rat hearts. We also demonstrate that iAng II is
correlated with the development of pathological condi-
tions associated with diabetes. Significantly, we observed
that blockade of the RAS by a renin inhibitor in diabetic
rats provided greater protection from oxidative stress and
cardiac fibrosis compared with inhibition with an AT
1
antagonist or ACE inhibitor.
We first described a physiologically relevant intracellu-
lar RAS in NRVMs (14), defined as the presence of the
precursor protein and enzymes and synthesis of Ang II
inside the cell and which was coupled to a biological
action (11). The regulation and separation of the intracel-
lular from the extracellular RAS becomes very obvious in
hyperglycemic conditions. High glucose promoted accu-
mulation of AGT, renin, and Ang II intracellularly, result-
ing in a dramatic rise in iAng II concentrations without
affecting extracellular Ang II levels (14). Similar to NRVMs,
the intracellular accumulation of RAS components and
iAng II synthesis have also been described in VSMCs and
renal mesangial cells in high-glucose culture conditions
(15,17,29). To extend the in vitro observations, we deter-
mined whether a similar activation of the intracellular RAS
occurs in adult diabetic animals. We observed a significant
increase in intracellular levels of AGT and renin and iAng
II synthesis in diabetic rat hearts, as determined by ELISA
and confocal immunocytochemistry. In a previous human
study, a threefold increase in Ang II staining was described
in hearts from diabetic compared with nondiabetic pa-
tients, which was enhanced an additional 2.5-fold in dia-
betic hypertensive patients (30). However, in that study, it
was not clear whether the increased Ang II staining
represented activation of an intracellular RAS or was due
to internalization of extracellularly synthesized Ang II. We
observed an increase in iAng II levels even in the presence
of AT
1
blockade with candesartan, which strongly sup-
ported activation of the intracellular RAS. The latter
conclusion was further strengthened by an observed in-
crease in intracellular staining for renin and AGT.
The observation of elevated Ang II levels after removal
of the high-glucose stimulus, suggested that iAng II was
highly stable. The reported half-life of Ang II in the heart
is 15 min in vivo and 30 min ex vivo (31). In this regard, it
is important to make a distinction between Ang II that was
internalized versus that which was synthesized intracellu-
larly, because the intracellular location is likely to be
different, which could substantially affect the half-life. The
half-life reported in the literature was for Ang II that was
internalized through AT
1
receptor (31), a major part of
which was likely targeted to lysosomes for degradation
(32). We measured Ang II that was synthesized intracellu-
larly, which is most likely to occur in organelles or at sites
that are not associated with protein degradation.
An interesting and therapeutically significant aspect of
the intracellular RAS is that high glucose–induced iAng II
synthesis in cardiac myocytes is catalyzed by chymase, not
ACE. Chymase levels are significantly elevated in NRVMs
after exposure to high glucose, whereas ACE levels remain
unchanged (14). Similarly, no change in gene expression of
ACE was observed in diabetic rat hearts (33). In the
above-referenced human study (30), diabetic patients who
had elevated iAng II levels were on ACE-inhibitor therapy.
In the latter case, a rise in Ang II levels could be attributed
to an “ACE-escape” phenomenon, which is believed to
occur after prolonged treatment with ACE inhibitors (34).
In the present study, the lack of reduction in iAng II levels
after only 1 week of ACE-inhibitor treatment strongly
suggests an ACE-independent mechanism of iAng II syn-
thesis, corroborating in vitro observations. Upregulation
of vascular chymase in diabetic patients and chymase-
mediated Ang II generation in human and rat VSMCs and
human mesangial cells has been previously reported
(15,17,18,35). Involvement of chymase further strengthens
the concept of an intracellular RAS in diabetes.
Renin inhibition by aliskiren completely prevented hy-
perglycemia-induced iAng II synthesis. The source of renin
in the heart has been an issue of debate (36). Circulating
prorenin levels are elevated severalfold in diabetes, which
B
AC
DEF
FIG. 5. Detection of cardiac fibrosis by Masson’s Trichrome staining in heart sections from control rats (A); diabetic rats (B); and diabetic rats
treated with insulin (C), aliskiren (D), candesartan (E), or benazepril (F). Representative images of five sections per heart and three hearts per
group were observed with a 40 objective. (Please see http://dx.doi.org/10.2337/db08-0805 for a high-quality digital representation of this figure.)
INTRACELLULAR Ang II IN DIABETES
3302 DIABETES, VOL. 57, DECEMBER 2008
might also contribute to cardiac levels of pro(renin) (37).
Cardiac myocytes have been shown to internalize and
activate prorenin, which could contribute to iAng II syn-
thesis (38,39). In addition, several reports have described
expression of renin by cardiac myocytes, fibroblasts, and
cardiac mast cells (13,40 42). Adult rat cardiac myocytes
have been shown to express an intracellular form of
prorenin, which lacked a portion of the preprofragment,
eliminating the need for proteolytic activation (43). We
have previously described a significant increase in intra-
cellular renin levels in NRVMs exposed to high-glucose
conditions (14). In the current study, we observed en-
hanced staining for renin in the heart of diabetic rats.
Thus, inhibition of cardiac iAng II synthesis by aliskiren
was consistent with these observations.
Although aliskiren is the most potent inhibitor of human
renin (half-maximal inhibitory concentration [IC
50
] 0.6
nmol/l), it inhibits rat renin at higher concentrations (IC
50
80 nmol/l) (44). We chose an aliskiren dose of 30 mg kg
1
day
1
based on significant blood pressure–lowering ef-
fects of this dose in spontaneously hypertensive rats (45).
Pharmacokinetic studies of aliskiren in Sprague-Dawley
rats (species used in this study) demonstrated that an oral
dose of 30 mg/kg resulted in an area under the curve of
3.06 1.8 mol l
1
h
1
, indicating sufficient drug in the
bloodstream (45). Several additional characteristics of
aliskiren could explain the observed effects in diabetic
rats. These include cellular uptake, accumulation on mul-
tiple dosing, longer half-life, and rapid binding to renin
with slow dissociation (46). We observed that neonatal rat
cardiac myocytes internalized aliskiren in a concentration-
dependent manner, levels of which were measured at 1,
24, and 48 h after addition to the culture medium. Maxi-
mum intracellular levels of aliskiren were observed at
24 h, which were not reduced even after 48 h (data not
shown). Aliskiren levels were measured by liquid chroma-
tography tandem mass spectrometry, as previously de-
scribed (47). Similarly, rats that were treated with
aliskiren at a dose of 10 mg kg
1
day
1
for 2 weeks,
showed a kidney-to-plasma ratio of aliskiren in the range
of 45 to 64 (47). The latter indicated extensive partitioning
of aliskiren to the kidneys, which localized to glomeruli
and the walls of small cortical arteries. Persistent renal
protective effects of aliskiren after discontinuation of
treatment also suggest slow clearance and accumulation
in tissues (48). The above findings suggest similar parti-
tioning of aliskiren in the heart, which might have resulted
in sufficiently high intracellular levels of aliskiren to inhibit
rat renin in our studies. Furthermore, tissue accumulation
of aliskiren may result in effects on target organs, even at
doses that do not affect blood pressure. This was evident
from a recent study wherein 2.5 mg/kg aliskiren did not
produce a statistically significant sustained reduction in
blood-pressure but reduced atherosclerotic lesion size
significantly in hypercholesterolemic Ldlr
/
mice (49).
Higher doses of aliskiren (up to 50 mg/kg) produced
AGEs
Glucose
GAD -3P
TCA cycle
Hyperglycemia
Hexosamine
Biosynthetic Pathway
O-glycosylation of
transcription factors
AGT
Renin
Chymase
AGT
Ang IAng II
Oxidative Stress
AT1 AGTAng IAng II
Renin
Chymase
ACE Renin
?
DAG PKC
Apoptosis
Fibrosis
?
FIG. 6. Schematic representation of the relationship between hyperglycemia, iAng II, and pathological effects. In hyperglycemia, there is an
increase in glucose oxidation through the tricarboxylic acid cycle in mitochondria, which results in enhanced generation of reactive oxygen
species. Overproduction of superoxide inhibits glyceraldehyde-3-phosphate dehydrogenase activity, resulting in an accumulation of upstream
metabolites of the glycolytic pathway. Increased levels of glyceraldehyde-3-phosphate (GAD-3P) cause activation of PKC isoforms through
diacylglycerol (DAG) production and synthesis of advanced glycation end products (AGEs). There is increased shuttling of glucose through the
hexosamine biosynthesis pathway, resulting in the modification of transcription factors through o-glycosylation. All of these products of
hyperglycemia, i.e., oxidative stress, AGEs, PKC, and o-glycosylation of transcription factors, activate expression of RAS components. Cardiac
myocytes synthesize and retain Ang II intracellularly in hyperglycemia, whereas cardiac fibroblasts increase both intra- and extracellular Ang II.
iAng II could directly increase oxidative stress and cellular apoptosis through unidentified mechanisms and/or could enhance expression of RAS
components through a positive feedback mechanism, resulting in enhanced extracellular Ang II levels as well, particularly via cardiac fibroblasts.
Extracellular Ang II in turn causes oxidative stress, cardiac myocyte apoptosis, and cardiac fibrosis through the AT
1
receptor. Interrupting this
cycle by blocking Ang II synthesis provides protection from hyperglycemia-induced pathological events. ACE inhibitors or ARBs would block only
the extracellular synthesis or actions, respectively, of Ang II; whereas a renin inhibitor would block both intra- and extracellular Ang II synthesis,
the latter providing an explanation for the more pronounced effects of aliskiren observed in this study.
V.P. SINGH AND ASSOCIATES
DIABETES, VOL. 57, DECEMBER 2008 3303
similar results. In the double human renin-AGT transgenic
rat model, a dose (of aliskiren) of 0.03 mg/kg, which did
not decrease blood pressure, reduced albuminuria and
cardiac hypertrophy (50).
iAng II has been shown to produce multiple biological
actions, including cardiac hypertrophy (11,19). Many of
the reported iAng II effects are not prevented by ARBs,
either due to limited cell permeability of these drugs or to
an AT
1
-independent mechanism of iAng II–mediated ef-
fects. We previously demonstrated that iAng II–induced
NRVM cell growth and cardiac hypertrophy was not
inhibited by ARBs (19). Proliferation of Chinese hamster
ovary cells, which are deficient in AT
1
receptor, demon-
strated that some of the effects of iAng II do not require
AT
1
receptor (22). In addition, enhanced transforming
growth factor-/smad signaling was reported in kidneys
from diabetic AT
1
-knockout mice (51). These findings,
together with the observation that iAng II synthesis is
chymase dependent, suggest that ARBs and ACE inhibi-
tors do not block the intracellular RAS, which is activated
in diabetes. A renin inhibitor prevents both intracellular
and extracellular Ang II synthesis (14) and thus may prove
more beneficial in diabetic conditions. Consistent with the
latter hypothesis, we observed that aliskiren was more
effective in preventing oxidative stress and cardiac fibrosis
compared with candesartan or benazepril. Effects of
aliskiren were unlikely to be mediated through Ang II–
independent mechanisms because aliskiren does not in-
hibit renin binding to the (pro)renin receptor and
extracellular signal–related kinase activation (8,47). The
effect of these drugs on cardiac myocyte apoptosis dem-
onstrated that aliskiren was significantly more beneficial
than benazepril.
Reversal of cardiac effects in diabetic animals by insulin
treatment indicates that the observed effects were due to
hyperglycemia. Although STZ-induced diabetes is repre-
sentative of type 1 diabetes, we also predict activation of
the intracellular RAS in type 2 diabetes. This is based on
our unpublished observations of no effect of insulin treat-
ment on high glucose–induced iAng II synthesis in NRVMs
and patients with type 2 diabetes, who showed enhanced
iAng II staining in the heart (30).
As depicted in Fig. 6, activation of the RAS appears to be
a major event in hyperglycemia as a result of increased
oxidative stress, increased protein kinase C (PKC) levels,
and/or increased activity of the hexosamine biosynthesis
pathway. iAng II could directly produce oxidative stress
and cellular apoptosis through unidentified mechanisms
and/or could enhance expression of RAS components
through a positive feedback mechanism (52), resulting in
enhanced extracellular Ang II levels as well, particularly
via cardiac fibroblasts (13). Extracellular Ang II in turn
causes oxidative stress, cardiac myocyte apoptosis, and
cardiac fibrosis through the AT
1
receptor. Interrupting this
cycle by blocking Ang II synthesis protects from hypergly-
cemia-induced pathological events. ACE inhibitors or
ARBs would block only the extracellular synthesis or
actions, respectively, of Ang II; whereas a renin inhibitor
would block both intra- and extracellular Ang II synthesis,
the latter providing an explanation for the more pro-
nounced effects of aliskiren observed in this study. In
diabetic heart, the source and target of Ang II could be
represented by multiple cell types. In addition to NRVMs,
we previously demonstrated that cardiac fibroblasts re-
spond to high glucose with enhanced activity of the RAS
and extracellular matrix production (13). Cardiac fibro-
blasts increase both intracellular and extracellular Ang II,
in contrast to cardiac myocytes, which demonstrate only
increased iAng II in high-glucose conditions. Additionally,
Ang II synthesis by cardiac fibroblasts, extracellular as
well as intracellular, is catalyzed by ACE (13). Thus, ACE
inhibitors would block Ang II synthesis by cardiac fibro-
blasts, and ARBs would block autocrine/paracrine effects
of extracellular Ang II. This explains the partial effects of
these agents in diabetic rats. However, these agents would
not block high glucose–stimulated iAng II synthesis or
intracellular actions in cardiac myocytes. In addition to a
direct effect on cardiac myocytes, iAng II synthesized in
cardiac myocytes would likely have indirect functional
effects on other cells by stimulating synthesis and release
of growth factors and cytokines from myocytes (53). Thus,
the intracellular RAS possibly explains progression from
microalbuminuria to proteinuria in diabetic patients on
ACE inhibitor therapy, the mechanism of resistance to
antihypertensive therapy in type 2 diabetes and higher
cardiovascular morbidity and mortality in hypertensive
patients with diabetes (10,54 –56). Although ARBs and
ACE inhibitors would provide some protection to the
cardiovascular system through positive hemodynamic ef-
fects, partial inhibition of the local RAS, or other non-
RAS–related mechanisms, such as an effect on peroxisome
proliferator–activated receptor-and the kallikrein-kinin
system (57–59); additional blockade of the intracellular
RAS using agents such as a renin inhibitor might prove
more beneficial in diabetes. Long-term studies that include
cardiac functional analysis will be necessary to validate
the above hypothesis.
ACKNOWLEDGMENTS
We thank the Texas A&M Health Science Center Micros-
copy Imaging Center for providing confocal microscopy
services. R.K. has received an American Heart Association,
Texas Affiliate, Beginning Grant-In-Aid.
REFERENCES
1. Paul M, Poyan Mehr A, Kreutz R: Physiology of local renin-angiotensin
systems. Physiol Rev 86:747– 803, 2006
2. Connelly KA, Boyle AJ, Kelly DJ: Angiotensin II and the cardiac compli-
cations of diabetes mellitus. Curr Pharm Des 13:2721–2729, 2007
3. Nguyen G, Delarue F, Berrou J, Rondeau E, Sraer JD: Specific receptor
binding of renin on human mesangial cells in culture increases plasmino-
gen activator inhibitor-1 antigen. Kidney Int 50:1897–1903, 1996
4. Nguyen G, Delarue F, Burckle C, Bouzhir L, Giller T, Sraer JD: Pivotal role
of the renin/prorenin receptor in angiotensin II production and cellular
responses to renin. J Clin Invest 109:1417–1427, 2002
5. Luetscher JA, Kraemer FB, Wilson DM, Schwartz HC, Bryer-Ash M:
Increased plasma inactive renin in diabetes mellitus: a marker of micro-
vascular complications. N Engl J Med 312:1412–1417, 1985
6. Kang JJ, Toma I, Sipos A, Meer EJ, Vargas SL, Peti-Peterdi J: The collecting
duct is the major source of prorenin in diabetes. Hypertension 51:1597–
1604, 2008
7. Saris JJ, ‘t Hoen PA, Garrelds IM, Dekkers DH, den Dunnen JT, Lamers JM,
Jan Danser AH: Prorenin induces intracellular signaling in cardiomyocytes
independently of angiotensin II. Hypertension 48:564 –571, 2006
8. Feldt S, Batenburg WW, Mazak I, Maschke U, Wellner M, Kvakan H,
Dechend R, Fiebeler A, Burckle C, Contrepas A, Jan Danser AH, Bader
M, Nguyen G, Luft FC, Muller DN: Prorenin and renin-induced extra-
cellular signal-regulated kinase 1/2 activation in monocytes is not
blocked by aliskiren or the handle-region peptide. Hypertension 51:
682– 688, 2008
9. Turnbull F, Neal B, Algert C, Chalmers J, Chapman N, Cutler J, Woodward
M, MacMahon S: Effects of different blood pressure-lowering regimens on
major cardiovascular events in individuals with and without diabetes
mellitus: results of prospectively designed overviews of randomized trials.
Arch Intern Med 165:1410 –1419, 2005
INTRACELLULAR Ang II IN DIABETES
3304 DIABETES, VOL. 57, DECEMBER 2008
10. Ficociello LH, Perkins BA, Silva KH, Finkelstein DM, Ignatowska-Swital-
ska H, Gaciong Z, Cupples LA, Aschengrau A, Warram JH, Krolewski AS:
Determinants of progression from microalbuminuria to proteinuria in
patients who have type 1 diabetes and are treated with angiotensin-
converting enzyme inhibitors. Clin J Am Soc Nephrol 2:461– 469, 2007
11. Kumar R, Singh VP, Baker KM: The intracellular renin-angiotensin system:
a new paradigm. Trends Endocrinol Metab 18:208 –214, 2007
12. Kumar R, Singh VP, Baker KM: The intracellular renin-angiotensin system:
implications in cardiovascular remodeling. Curr Opin Nephrol Hypertens
17:168 –173, 2008
13. Singh VP, Baker KM, Kumar R: Activation of the intracellular renin-
angiotensin system in cardiac fibroblasts by high glucose: role in extracel-
lular matrix production. Am J Physiol Heart Circ Physiol 294:H1675–
H1684, 2008
14. Singh VP, Le B, Bhat VB, Baker KM, Kumar R: High glucose induced
regulation of intracellular angiotensin II synthesis and nuclear redistribu-
tion in cardiac myocytes. Am J Physiol Heart Circ Physiol 293:H939 –
H948, 2007
15. Lavrentyev EN, Estes AM, Malik KU: Mechanism of high glucose induced
angiotensin II production in rat vascular smooth muscle cells. Circ Res
101:455– 464, 2007
16. Singh R, Leehey DJ: Effect of ACE inhibitors on angiotensin II in rat
mesangial cells cultured in high glucose. Biochem Biophys Res Commun
357:1040 –1045, 2007
17. Cristovam PC, Arnoni CP, de Andrade MC, Casarini DE, Pereira LG, Schor
N, Boim MA: ACE- and chymase-dependent angiotensin II generation in
normal and glucose-stimulated human mesangial cells. Exp Biol Med
233:1035–1043, 2008
18. Koka V, Wang W, Huang XR, Kim-Mitsuyama S, Truong LD, Lan HY:
Advanced glycation end products activate a chymase-dependent angioten-
sin II-generating pathway in diabetic complications. Circulation 113:1353–
1360, 2006
19. Baker KM, Chernin MI, Schreiber T, Sanghi S, Haiderzaidi S, Booz GW,
Dostal DE, Kumar R: Evidence of a novel intracrine mechanism in
angiotensin II-induced cardiac hypertrophy. Regul Pept 120:5–13, 2004
20. Cook JL, Zhang Z, Re RN: In vitro evidence for an intracellular site of
angiotensin action. Circ Res 89:1138 –1146, 2001
21. Filipeanu CM, Brailoiu E, Kok JW, Henning RH, De Zeeuw D, Nelemans SA:
Intracellular angiotensin II elicits Ca2increases in A7r5 vascular smooth
muscle cells. Eur J Pharmacol 420:9 –18, 2001
22. Baker KM, Kumar R: Intracellular angiotensin II induces cell proliferation
independent of AT1 receptor. Am J Physiol Cell Physiol 291:C995–C1001,
2006
23. Weber MA, Giles TD: Inhibiting the renin-angiotensin system to prevent
cardiovascular diseases: do we need a more comprehensive strategy? Rev
Cardiovasc Med 7:45–54, 2006
24. Kajstura J, Fiordaliso F, Andreoli AM, Li B, Chimenti S, Medow MS, Limana
F, Nadal-Ginard B, Leri A, Anversa P: IGF-1 overexpression inhibits the
development of diabetic cardiomyopathy and angiotensin II-mediated
oxidative stress. Diabetes 50:1414 –1424, 2001
25. Raimondi L, De Paoli P, Mannucci E, Lonardo G, Sartiani L, Banchelli G,
Pirisino R, Mugelli A, Cerbai E: Restoration of cardiomyocyte functional
properties by angiotensin II receptor blockade in diabetic rats. Diabetes
53:1927–1933, 2004
26. Nussberger J, Wuerzner G, Jensen C, Brunner HR: Angiotensin II suppres-
sion in humans by the orally active renin inhibitor Aliskiren (SPP100):
comparison with enalapril. Hypertension 39:E1–E8, 2002
27. Delacretaz E, Nussberger J, Biollaz J, Waeber B, Brunner HR: Character-
ization of the angiotensin II receptor antagonist TCV-116 in healthy
volunteers. Hypertension 25:14 –21, 1995
28. Juillerat L, Nussberger J, Menard J, Mooser V, Christen Y, Waeber B, Graf
P, Brunner HR: Determinants of angiotensin II generation during convert-
ing enzyme inhibition. Hypertension 16:564 –572, 1990
29. Vidotti DB, Casarini DE, Cristovam PC, Leite CA, Schor N, Boim MA: High
glucose concentration stimulates intracellular renin activity and angioten-
sin II generation in rat mesangial cells. Am J Physiol Renal Physiol
286:F1039 –F1045, 2004
30. Frustaci A, Kajstura J, Chimenti C, Jakoniuk I, Leri A, Maseri A, Nadal-
Ginard B, Anversa P: Myocardial cell death in human diabetes. Circ Res
87:1123–1132, 2000
31. van Kats JP, de Lannoy LM, Jan Danser AH, van Meegen JR, Verdouw PD,
Schalekamp MA: Angiotensin II type 1 (AT1) receptor-mediated accumu-
lation of angiotensin II in tissues and its intracellular half-life in vivo.
Hypertension 30:42– 49, 1997
32. Bianchi C, Gutkowska J, De Lean A, Ballak M, Anand-Srivastava MB,
Genest J, Cantin M: Fate of [125I]angiotensin II in adrenal zona glomeru-
losa cells. Endocrinology 118:2605–2607, 1986
33. Fiordaliso F, Li B, Latini R, Sonnenblick EH, Anversa P, Leri A, Kajstura J:
Myocyte death in streptozotocin-induced diabetes in rats in angiotensin II-
dependent. Lab Invest 80:513–527, 2000
34. van de Wal RM, Plokker HW, Lok DJ, Boomsma F, van der Horst FA, van
Veldhuisen DJ, van Gilst WH, Voors AA: Determinants of increased
angiotensin II levels in severe chronic heart failure patients despite ACE
inhibition. Int J Cardiol 106:367–372, 2006
35. Li M, Liu K, Michalicek J, Angus JA, Hunt JE, Dell’Italia LJ, Feneley MP,
Graham RM, Husain A: Involvement of chymase-mediated angiotensin II
generation in blood pressure regulation. J Clin Invest 114:112–120, 2004
36. Krop M, Danser AH: Circulating versus tissue renin-angiotensin system: on
the origin of (pro)renin. Curr Hypertens Rep 10:112–118, 2008
37. Franken AA, Derkx FH, Man in’t Veld AJ, Hop WC, van Rens GH,
Peperkamp E, de Jong PT, Schalekamp MA: High plasma prorenin in
diabetes mellitus and its correlation with some complications. J Clin
Endocrinol Metab 71:1008 –1015, 1990
38. van Kesteren CA, Danser AH, Derkx FH, Dekkers DH, Lamers JM, Saxena
PR, Schalekamp MA: Mannose 6-phosphate receptor-mediated internaliza-
tion and activation of prorenin by cardiac cells. Hypertension 30:1389 –
1396, 1997
39. Peters J, Farrenkopf R, Clausmeyer S, Zimmer J, Kantachuvesiri S, Sharp
MGF, Mullins JJ: Functional significance of prorenin internalization in the
rat heart. Circ Res 90:1135–1141, 2002
40. Peters J: Secretory and cytosolic (pro)renin in kidney, heart, and adrenal
gland. J Mol Med 86:711–714, 2008
41. Sanghi S, Kumar R, Smith M, Baker KM, Dostal DE: Activation of protein
kinase A by atrial natriuretic peptide in neonatal rat cardiac fibroblasts:
role in regulation of the local renin-angiotensin system. Regul Pept
132:1– 8, 2005
42. Mackins CJ, Kano S, Seyedi N, Schafer U, Reid AC, Machida T, Silver RB,
Levi R: Cardiac mast cell-derived renin promotes local angiotensin forma-
tion, norepinephrine release, and arrhythmias in ischemia/reperfusion.
J Clin Invest 116:1063–1070, 2006
43. Peters J, Clausmeyer S: Intracellular sorting of renin: cell type specific
differences and their consequences. J Mol Cell Cardiol 34:1561–1568,
2002
44. Wood JM, Maibaum J, Rahuel J, Grutter MG, Cohen NC, Rasetti V, Ruger
H, Goschke R, Stutz S, Fuhrer W, Schilling W, Rigollier P, Yamaguchi Y,
Cumin F, Baum HP, Schnell CR, Herold P, Mah R, Jensen C, O’Brien E,
Stanton A, Bedigian MP: Structure-based design of aliskiren, a novel orally
effective renin inhibitor. Biochem Biophys Res Commun 308:698–705,
2003
45. Wood JM, Schnell CR, Cumin F, Menard J, Webb RL: Aliskiren, a novel,
orally effective renin inhibitor, lowers blood pressure in marmosets and
spontaneously hypertensive rats. J Hypertens 23:417– 426, 2005
46. Hong Y, Dingemanse J, Mager DE: Pharmacokinetic/pharmacodynamic
modeling of renin biomarkers in subjects treated with the renin inhibitor
aliskiren. Clin Pharmacol Ther 84:136 –143, 2008
47. Feldman DL, Jin L, Xuan H, Contrepas A, Zhou Y, Webb RL, Mueller DN,
Feldt S, Cumin F, Maniara W, Persohn E, Schuetz H, Jan Danser AH,
Nguyen G: Effects of aliskiren on blood pressure, albuminuria, and
(pro)renin receptor expression in diabetic TG(mRen-2)27 rats. Hyperten-
sion 52:130 –136, 2008
48. Feldman DL, Jin L, Miserindino-Moltini R, Xuan H, Luft FC, Muller DN:
P-613: Aliskiren, a human renin inhibitor induces persistent renoprotection
comparable to ACE inhibition in double transgenic rats (dTGR). Am J
Hypertens 18:230A, 2005
49. Lu H, Rateri DL, Feldman DL, Jr RJ, Fukamizu A, Ishida J, Oesterling EG,
Cassis LA, Daugherty A: Renin inhibition reduces hypercholesterolemia-
induced atherosclerosis in mice. J Clin Invest 118:984 –993, 2008
50. Dechend R, Shagdarsuren E, Gratze P, Fiebeler A, Pilz B, Meiners S, Derer
W, Feldman DL, Webb R, Muller DN: Low-dose renin inhibitor and
low-dose AT(1)-receptor blocker therapy ameliorate target-organ damage
in rats harbouring human renin and angiotensinogen genes. J Renin
Angiotensin Aldosterone Syst 8:81– 84, 2007
51. Okazaki Y, Yamasaki Y, Uchida HA, Okamoto K, Satoh M, Maruyama K,
Maeshima Y, Sugiyama H, Sugaya T, Kashihara N, Makino H: Enhanced
TGF-beta/Smad signaling in the early stage of diabetic nephropathy is
independent of the AT1a receptor. Clin Exp Nephrol 11:77– 87, 2007
52. Singh VP, Baker KM, Kumar R: Intracellular angiotensin II is a positive
regulator of the cardiac renin-angiotensin system (Abstract). Hyperten-
sion 52:e91, 2008
53. Westermann D, Rutschow S, Van Linthout S, Linderer A, Bucker-Gartner C,
V.P. SINGH AND ASSOCIATES
DIABETES, VOL. 57, DECEMBER 2008 3305
Sobirey M, Riad A, Pauschinger M, Schultheiss HP, Tschope C: Inhibition
of p38 mitogen-activated protein kinase attenuates left ventricular dys-
function by mediating pro-inflammatory cardiac cytokine levels in a mouse
model of diabetes mellitus. Diabetologia 49:2507–2513, 2006
54. Eguchi K, Pickering TG, Kario K: Why is blood pressure so hard to control
in patients with type 2 diabetes? J Cardiometab Syndr 2:114 –118, 2007
55. Aksnes TA, Kjeldsen SE, Rostrup M, Omvik P, Hua TA, Julius S: Impact of
new-onset diabetes mellitus on cardiac outcomes in the Valsartan Antihy-
pertensive Long-term Use Evaluation (VALUE) trial population. Hyperten-
sion 50:467– 473, 2007
56. Okin PM, Devereux RB, Gerdts E, Snapinn SM, Harris KE, Jern S, Kjeldsen
SE, Julius S, Edelman JM, Lindholm LH, Dahlof B: Impact of diabetes
mellitus on regression of electrocardiographic left ventricular hypertrophy
and the prediction of outcome during antihypertensive therapy: the
Losartan Intervention For Endpoint (LIFE) Reduction in Hypertension
Study. Circulation 113:1588 –1596, 2006
57. Schupp M, Janke J, Clasen R, Unger T, Kintscher U: Angiotensin type 1
receptor blockers induce peroxisome proliferator-activated receptor-
gamma activity. Circulation 109:2054 –2057, 2004
58. Watanabe T, Suzuki J, Yamawaki H, Sharma VK, Sheu SS, Berk BC:
Losartan metabolite EXP3179 activates Akt and endothelial nitric oxide
synthase via vascular endothelial growth factor receptor-2 in endothelial
cells: angiotensin II type 1 receptor-independent effects of EXP3179.
Circulation 112:1798 –1805, 2005
59. Yoshiyama M, Nakamura Y, Omura T, Izumi Y, Matsumoto R, Oda S,
Takeuchi K, Kim S, Iwao H, Yoshikawa J: Angiotensin converting enzyme
inhibitor prevents left ventricular remodelling after myocardial infarction
in angiotensin II type 1 receptor knockout mice. Heart 91:1080 –1085, 2005
INTRACELLULAR Ang II IN DIABETES
3306 DIABETES, VOL. 57, DECEMBER 2008

Supplementary resource (1)

... Activation of the adrenergic system increases β-adrenergic expression and signaling, promoting myocyte hypertrophy, interstitial fibrosis, myocyte apoptosis, and contractile dysfunction [78]. In experimental models of T1D, an elevation in angiotensin-II receptor density and synthesis has been observed [57,79]. ...
... CAN is known to occur in individuals with T1D, correlating with increased CVD and mortality [79,80]. It is suggested that cardiac neuropathy may affect up to 40% of individuals with T1D [82,83]. ...
Article
Full-text available
Heart failure (HF) is increasing at an alarming rate, primary due to the rising in aging, obesity and diabetes. Notably, individuals with type 1 diabetes (T1D) face a significantly elevated risk of HF, leading to more hospitalizations and increased case fatality rates. Several risk factors contribute to HF in T1D, including poor glycemic control, female gender, smoking, hypertension, elevated BMI, and albuminuria. However, early and intensive glycemic control can mitigate the long-term risk of HF in individuals with T1D. The pathophysiology of diabetes-associated HF is complex and multifactorial, and the underlying mechanisms in T1D remain incompletely elucidated. In terms of treatment, much of the evidence comes from type 2 diabetes (T2D) populations, so applying it to T1D requires caution. Sodium-glucose cotransporter 2 inhibitors have shown benefits in HF outcomes, even in non-diabetic populations. However, most of the information about HF and the evidence from cardiovascular safety trials related to glucose lowering medications refer to T2D. Glycemic control is key, but the link between hypoglycemia and HF hospitalization risk requires further study. Glycemic variability, common in T1D, is an independent HF risk factor. Technological advances offer the potential to improve glycemic control, including glycemic variability, and may play a role in preventing HF. In summary, HF in T1D is a complex challenge with unique dimensions. This review focuses on HF in individuals with T1D, exploring its epidemiology, risk factors, pathophysiology, diagnosis and treatment, which is crucial for developing tailored prevention and management strategies for this population.
... Then, for investigating the specific effects of the five main active compounds, Ang II-induced cardiomyocyte hypertrophy model was exploited. Ang II induces an inflammatory phenotype in cardiomyocytes,46 and leads to cellular hypertrophy47 and increased deposition of matrix proteins.48 Furthermore, inhibiting Ang II has been shown to prevent cardiac hypertrophy and fibrosis.49 ...
Article
Full-text available
Heart failure is a leading cause of death in the elderly. Traditional Chinese medicine, a verified alternative therapeutic regimen, has been used to treat heart failure, which is less expensive and has fewer adverse effects. In this study, a total of 15 active ingredients of Astragalus membranaceus (Huangqi, HQ) were obtained; among them, Isorhamnetin, Quercetin, Calycosin, Formononetin, and Kaempferol were found to be linked to heart failure. Ang II significantly enlarged the cell size of cardiomyocytes, which could be partially reduced by Quercetin, Isorhamnetin, Calycosin, Kaempferol, or Formononetin. Ang II significantly up‐regulated ANP, BNP, β‐MHC, and CTGF expressions, whereas Quercetin, Isorhamnetin, Calycosin, Kaempferol or Formononetin treatment partially downregulated ANP, BNP, β‐MHC and CTGF expressions. Five active ingredients of HQ attenuated inflammation in Ang II‐induced cardiomyocytes by inhibiting the levels of TNF‐α, IL‐1β, IL‐18 and IL‐6. Molecular docking shows Isorhamnetin, Quercetin, Calycosin, Formononetin and Kaempferol can bind with its target protein ESR1 in a good bond by intermolecular force. Quercetin, Calycosin, Kaempferol or Formononetin treatment promoted the expression levels of ESR1 and phosphorylated ESR1 in Ang II‐stimulated cardiomyocytes; however, Isorhamnetin treatment had no effect on ESR1 and phosphorylated ESR1 expression levels. In conclusion, our results comprehensively illustrated the bioactives, potential targets, and molecular mechanism of HQ against heart failure. Isorhamnetin, Quercetin, Calycosin, Formononetin and Kaempferol might be the primary active ingredients of HQ, dominating its cardioprotective effects against heart failure through regulating ESR1 expression, which provided a basis for the clinical application of HQ to regulate cardiac hypertrophy and heart failure.
... In T1D subjects, there is an increased activity of the renin-angiotensin-aldosterone system (RAAS) that causes an increase in cardiac fibrotic remodeling; in a 2008 study by Sing et al., T1D was induced in rats by injection of streptozotocin: after 1 week of hyperglycemia, a significant increase in angiotensin 2 levels was detected in streptozotocintreated rats, which in turn led to increased cardiomyocyte apoptosis and fibrosis [57]. Ultimately, diabetic cardiomyopathy led to cardiac remodeling responsible for diastolic dysfunction and increased the left atrial size, resulting in atrial fibrillation [5]. ...
Article
Full-text available
Heart failure (HF) management in type 1 diabetes (T1D) is particularly challenging due to its increased prevalence and the associated risks of hospitalization and mortality, driven by diabetic cardiomyopathy. Sodium–glucose cotransporter-2 inhibitors (SGLT2-is) offer a promising avenue for treating HF, specifically the preserved ejection fraction variant most common in T1D, but their utility is hampered by the risk of euglycemic diabetic ketoacidosis (DKA). This review investigates the potential of SGLT2-is in T1D HF management alongside emergent Continuous Ketone Monitoring (CKM) technology as a means to mitigate DKA risk through a comprehensive analysis of clinical trials, observational studies, and reviews. The evidence suggests that SGLT2-is significantly reduce HF hospitalization and enhance cardiovascular outcomes. However, their application in T1D patients remains limited due to DKA concerns. CKM technology emerges as a crucial tool in this context, offering real-time monitoring of ketone levels, which enables the safe incorporation of SGLT2-is into treatment regimes by allowing for early detection and intervention in the development of ketosis. The synergy between SGLT2-is and CKM has the potential to revolutionize HF treatment in T1D, promising improved patient safety, quality of life, and reduced HF-related morbidity and mortality. Future research should aim to employ clinical trials directly assessing this integrated approach, potentially guiding new management protocols for HF in T1D.
... In T1D subjects there is an increased activity of the Renin-Angiotensin-Aldosterone system (RAAS) that causes an increase in cardiac fibrotic remodeling; in a 2008 study by Sing et al. T1D was induced in rats by injection of streptozotocin: after 1 week of hyperglycemia, a significant increase in angiotensin 2 levels was detected in streptozotocin-treated rats, which in turn led to increased cardiomyocyte apoptosis and fibrosis [54]. Ultimately, diabetic cardiomyopathy led to cardiac remodeling responsible for diastolic dysfunction and increased left atrial size, resulting in atrial fibrillation [4]. ...
Preprint
Full-text available
Heart failure (HF) management in type 1 diabetes (T1D) is particularly challenging due to its increased prevalence and the associated risks of hospitalization and mortality, driven by diabetic cardiomyopathy. Sodium-glucose cotransporter-2 inhibitors (SGLT2-i) offer a promising avenue for treating HF, specifically the preserved ejection fraction variant most common in T1D, but their utility is hampered by the risk of euglycemic diabetic ketoacidosis (DKA). This review investigates the potential of SGLT2-i in T1D HF management, alongside the emergent Continuous Ketone Monitoring (CKM) technology as a means to mitigate DKA risk, through a comprehensive analysis of clinical trials, observational studies, and reviews. The evidence suggests SGLT2-i significantly reduce HF hospitalization and enhance cardiovascular outcomes. However, their application in T1D patients remains limited due to DKA concerns. CKM technology emerges as a crucial tool in this context, offering real-time monitoring of ketone levels, which enables the safe incorporation of SGLT2-i into treatment regimes by allowing for early detection and intervention in the development of ketosis. The synergy between SGLT2-i and CKM has the potential to revolutionize HF treatment in T1D, promising improved patient safety, quality of life, and reduced HF-related morbidity and mortality. Future research should aim at clinical trials directly assessing this integrated approach, potentially guiding new management protocols for HF in T1D.
... Hyperglycemia-induced AngII and aldosterone activity promote the deposition of ECM proteins and extensive myocardial fibrosis, leading to cardiac remodeling and impaired function [47][48][49]. Regarding the antifibrotic effects of RAASis, previous preclinical studies in STZ-induced diabetic models have demonstrated the attenuations in myocardial interstitial fibrosis [50][51][52]. However, these studies used regular or high doses of RAASis [53][54][55]. ...
Article
Full-text available
Diabetic cardiovascular complications are associated with up to 50% mortality, and current therapies are not effective enough. Renin–angiotensin–aldosterone system inhibitors (RAASis) are the standard of care for diabetic patients with hypertension and albuminuria. Based on our previous studies reporting the renoprotective effects of low-dose RAASis, here, we hypothesized that low-dose RAASi treatment has cardioprotective and antifibrotic benefits in type 1 diabetes mellitus (T1DM). After five weeks of T1DM, adult male Wistar rats received low doses of ramipril, losartan, or eplerenone for two weeks. Heart rate, blood pressure, and pulse wave velocity (PWV) were recorded. Aortic intima–media thickness (IMT), collagen accumulation, and myocardial fibrosis were assessed. All RAASis reduced PWV elevation, prevented the progression of myocardial fibrosis, and normalized B-type natriuretic peptide, troponin I, and fibroblast growth factor 23 levels without affecting blood pressure. Interestingly, only eplerenone reversed the decline in Klotho levels and reduced IMT and fibrosis in the media of the aorta. Our comparative analysis suggests that mineralocorticoid receptor antagonists, particularly eplerenone, may offer superior efficacy in halting both the arterial and the myocardial injuries in T1DM compared to angiotensin-converting enzyme inhibitors or angiotensin II type 1 receptor blockers.
... Experimental proof indicates that variable separate modulators might further facilitating DM correlated fibrosis io neurohumoral factors, inflammatory cytokines, growth factors as well as endothelin -1, adipokines along with ROS (Kaur et al., 2020). Angiotensin converting enzyme (ACE2) hampering has been displayed to be correlated with decreased collagen laying down along with perivascular fibrosis in various rat models of DbCM (Singh et al., 2008).This pointed that enhanced renin-angiotensin--aldosterone -system (RAAS) activity found in diabetic myocardium might be implicated in the pathogenesis of DbCM (Senador et al., 2009). ...
Article
Full-text available
In case of diabetic patients cardiomyopathy is a significant etiology of heart failure (HF); howeverno clarification exists regarding its pathophysiology. Myocardial hypertrophy as well as diastolic impairment are the logo for Diabetic cardiomyopathy (DbCM), whereas systolic function gets impacted in the latter phase of this disease. Previously we had reviewed Diabetic cardiomyopathy (DbCM with emphasis on epigenetic alterations ,miR changes apart from adipocyte impairment in HF induction, role of SGLT2 hampering agents in beneficial Cardiovascular Outcome Trials (CVOT's)and renal benefits, role of sirtuins in macrophage Polarization for Diabesity therapy .Here our aim was to posit the pathophysiological mode implicated in myocardial hypertrophy and escalated myocardial stiffness resulting in diastolic impairment .This myocardial stiffness takes place from cellular and extracellular matrix(ECM) stiffness &cell-matrix crosstalk. Escalated inherent cardiomyocytes stiffness plausibly is the maximum significant cause responsible for myocardial stiffness. It leads to dysfunctional cardiomyocytes cytoskeleton stiffness. Various other modes are implicatedbyT2DM; particularly having significant influence on myocardial stiffening i.e dysfunctional nitric oxide (NO), coronary microvascular impairment, escalated inflammation, as well as Oxidative stress(OS), myocardial Sodium-glucose specific cotransporter 2(SGLT2) modulated actions. Greater insight aids in planning better therapy.Anti diabeticagents, SGLT2 hampering agents have revealed newer ways of benefit ,like sirtuins action , upregulation of nutrient deprivation signaling in addition to downregulation of nutrient surplus signaling, escalated expression &activity of AMP-activated protein kinase(AMPK), Sirtuins (SIRT1), SIRT3 andSIRT6, &Peroxisome Proliferator Activated Receptor γCoactivator-1α(PGC-1α) ,reduction in activation of mammalian target of rapamycin inhibitors (mTOR). Recently Ghosh et al. (2023),advocated targeting sarcoplasmic / cytosolic endoplasmic reticulum Ca 2+-ATPase(SERCA2) ,use of imeglimin which escalates insulin action& along with reverses Pancreatic βcells impairment ,use of anti oxidants, angiotensin receptor neprilysin hampering agents(ARNi),NO stimulating agents. More work is being done regarding combination of empagliflozin with linagliptin &others like semaglutide with .Gradually we will get answers by getting more insight.
Article
The placenta is a temporary organ that provides communication between the mother and fetus. Maternal diabetes and abnormal placental angiogenesis may be linked. We investigated the angiogenesis mechanism resulting from VEGF and glucose stimulation in PECs obtained from human term placenta. Immunohistochemistry was performed to characterize PECs obtained from human term placenta. D-glucose was added to the medium containing PECs to establish normoglycemic and hyperglycemic conditions. The expression levels of VEGF, VEGFR-1 and VEGFR-2 genes and proteins in PECs from the control and experimental groups were analyzed by RT-PCR and Western blotting, respectively. With 48-hours incubation, gene expressions increased due to hyperglycemia, while protein levels increased due to the combined effect of VEGF and hyperglycemia. While VEGFR-2 gene expression and protein amounts increased in 24-hours due to the combined effect of VEGF and hyperglycemia, the effect of VEGF stimulation and glucose level on VEGFR-2 decreased in 48-hour incubation with time. VEGF, VEGFR-1 and VEGFR-2 genes and proteins were affected by hyperglycemic conditions in PECs. Hyperglycemia occurring in various conditions such as gestational diabetes mellitus and diabetes mellitus may affect VEGF, VEGFR-1 and VEGFR-2 genes and proteins of PECs derived from human term placenta.
Article
Full-text available
Background and Purpose: The mechanistic target of rapamycin (mTOR) protein complex increases in response to resistance training with a histological approach and decreases in aerobic training with a metabolic approach. These changes can be discussed in the conditions of type 2 diabetes (T2DM) and with exercise. Therefore, taking into account exercises such as high-intensity interval training (HIIT) with aerobic and anaerobic characteristics and on the other hand T2DM intervention can depict the possible interaction of mTOR with glycemic and histopathological indicators of the cardiac. So, the present study was conducted to determine the effect of eight weeks of HIIT on p-mTOR, T-mTOR and fibrosis of cardiac tissue and insulin resistance on diabetic rats through STZ injection. Materials and Methods: Twenty male adult white Wistar rats were divided into four groups (N=5): healthy control, diabetic control, healthy workout and diabetic workout. The HIIT protocol was carried out five sessions a week for eight weeks on a treadmill. One-way analysis of variance was used for statistical analysis at a significance level of P < 0.05. Results: The results showed that eight weeks of HIIT did not lead to significant changes in the amount of p-mTOR and T-mTOR in the cardiac tissue of rats with T2DM (P > 0.05); However, it caused a significant decrease in insulin resistance (31.2% compared to the diabetic control group) and cardiac tissue fibrosis (51.6% compared to the diabetic control group) in rats with T2DM (P < 0.05). Conclusion: According to the results, it seems that eight weeks of HIIT is enough to observe the increase in amount of cardiac tissue mTOR in healthy rats, but it is insufficient in rats with T2DM. Also, it is possible that the reduction of cardiac tissue fibrosis in rats with T2DM, at least during eight weeks of HITT, is independent of cardiac tissue mTOR.
Chapter
The unprecedented increase in the prevalence of diabetes mellitus (DM) coupled with its impact on cardiovascular disease makes it a dominant public health problem of the first quarter of this century. Multiple factors lead to the activation of the renin–angiotensin–aldosterone system (RAAS) in diabetes. Experimental models also show that increases in AT1 receptor expression, changes in post-receptor signaling mechanisms, and/or a reduction in cellular signals that suppress AT1 responses also lead to an increase in the sensitivity of the vasculature to RAAS. This results in a propensity to develop premature and accelerated atherosclerosis in DM that leads to considerable short- and long-term morbidity and mortality. These changes are amplified by hypertension and kidney disease. Compelling evidence from clinical trials show that RAAS inhibitors reduce cardiovascular events and mortality related to acute myocardial infarction (MI) and heart failure among patients with diabetes. Inhibition of the RAAS system by either ACE inhibition or AT1 receptor antagonism can also increase insulin sensitivity and glucose utilization. Multiple large prospective trials involving ACE inhibitors or ARBs have also reported an unexpected reduction in the development of new type 2 diabetes mellitus in patients treated with these agents. Further studies are needed to characterize “crosstalk” within the RAAS and to determine whether targeting additional components in this system might provide new therapeutic opportunities.KeywordsRenin–angiotensin–aldosterone systemDiabetes mellitusAngiotensin II sensitivityAtherogenesisCardiovascular disease, Myocardial infarctionHeart failureInsulin sensitivityNew-onset diabetes
Article
Catheter ablation is an effective and durable treatment option for patients with atrial fibrillation (AF). Ablation outcomes vary widely, with optimal results in patients with paroxysmal AF and diminishing results in patients with persistent or long-standing persistent AF. A number of clinical factors including obesity, hypertension, diabetes, obstructive sleep apnea, and alcohol use contribute to AF recurrence following ablation, likely through modulation of the atrial electro-anatomic substrate. In this article, we review the clinical risk factors and the electro-anatomic features that contribute to AF recurrence in patients undergoing ablation for AF.
Article
Binding and internalization of [¹²⁵I]angiotensin II (AII) were studied by morphological and biochemical methods in rats in vivo. Light microscope radioautography demonstrated that [²⁵I]AII binds specifically to adrenal zona glomerulosa (ZG) cells. Ultrastructural radioautographic analysis revealed that [¹²⁵I]AII binds to the cell surface, clusters in coated pits, is internalized in coated vesicles, and is transported by receptosomes to lysosomes in less than 20 min. Biochemical analysis revealed that as much as 40% of the adrenal radioactive uptake behaves as native [¹²⁵I]AII as shown by electrophoresis, immunoprecipitation and radioligand binding studies. These results indicate that the effects of All on the secretion of aldosterone by ZG cells are mediated by cell surface phenomena and not by binding to intracellular organelles involved in steroidogenesis. They also indicate that the half-life of All bound to receptors and internalized seems to be much longer (min) than in the systemic circulation (sec).
Article
Am J Hypertens (2005) 18, 230A–230A; doi:10.1016/j.amjhyper.2005.03.630 P-613: Aliskiren, a human renin inhibitor induces persistent renoprotection comparable to ACE inhibition in double transgenic rats (dTGR) David L. Feldman1, Liang Jin1, Rocca Miserindino-Moltini1, Hong Xuan1, Friedrich C. Luft1 and Dominik N. Müller11Cardiovascular Disease Pharmacology, Novartis Institute for Biomedical Research, E. Hanover, NJ; Franz Volhard Clinic, Medical Faculty of the Charité, Berlin, Germany.
Article
The reaction of the renin-angiotensin system to acute angiotensin converting enzyme inhibition was investigated in a single-blind, crossover study in nine normal volunteers receiving two out of three regimens in random order: the new converting enzyme inhibitor benazepril (20 mg once or 5 mg four times at 6-hour intervals) or enalapril (20 mg). Plasma converting enzyme activity, drug levels, angiotensin I and angiotensin II, active renin, and aldosterone were measured before and 1-4 hours and 14-30 hours after drug intake. Baseline in vitro plasma converting enzyme activity was 97 +/- 15 nmol/ml/min (mean +/- SD) when Hip-Gly-Gly was used as substrate, but with carbobenzoxy-Phe-His-Leu (Z-Phe-His-Leu) or angiotensin I as substrate it was only 20 +/- 4 and 1.7 +/- 0.3 nmol/ml/min, respectively. Discriminating power at peak converting enzyme inhibition was enhanced with the two latter substrates. In vivo converting enzyme activity was estimated by the plasma angiotensin II/angiotensin I ratio, which correlated well with in vitro converting enzyme activity using Z-Phe-His-Leu as substrate (r = 0.76, n = 252). Angiotensin II levels returned to baseline less than 24 hours after drug administration, whereas in vitro and in vivo converting enzyme activity remained considerably inhibited and active renin together with angiotensin I levels were still elevated. A close linear relation was found between plasma angiotensin II and the angiotensin I/drug level ratio (r = 0.91 for benazeprilat and r = 0.88 for enalaprilat, p less than 0.001). Thus, plasma angiotensin II truly reflects the resetting of the renin-angiotensin system at any degree of converting enzyme inhibition. The ratio of plasma angiotensin II to angiotensin I represents converting enzyme inhibition more accurately than in vitro assays, which vary considerably depending on substrates and assay conditions used.
Article
Plasma prorenin is abnormally high, whereas renin is normal or even low, in many patients with long-standing diabetes mellitus complicated by microvascular disease. Nephropathy or autonomic neuropathy has been put forward as a cause. We found that in 223 consecutive diabetics prorenin correlated positively with serum creatinine, the presence of macroalbuminuria (greater than 250 mg/L), and the presence of diabetic retinopathy, particularly the proliferative type. This correlation did not depend on the presence of neuropathy or whether the patient was receiving insulin. It was also independent of sex, age, duration of diabetes, blood pressure, and blood levels of glucose and hemoglobin-A1c. The association between elevated prorenin and retinopathy remained significant after adjustment for creatinine and the presence of macroalbuminuria. Of the whole group of diabetics 94 consecutive patients were assessed for the presence of microalbuminuria (30-300 mg/24 h). Independently of the presence of micro- or macroalbuminuria, the mean level of prorenin was not above normal in the patients without retinopathy and was 2-3 times normal in those with proliferative retinopathy. Thus, retinopathy appears to be a more important determinant of abnormally high prorenin than nephropathy. In addition, the renal vein to artery ratio of prorenin in 7 diabetics with both advanced nephropathy and proliferative retinopathy was not elevated, despite the high peripheral venous prorenin level and the impaired renal perfusion. Thus, the abnormally high prorenin level in these patients could not be explained by abnormal secretion by the kidneys. Finally, prorenin was not high in 16 nondiabetics with loss of sympathetic activity due to chronic autonomic neuropathy, which indicates that in the absence of diabetes, this type of autonomic failure is not sufficient to cause the high prorenin levels seen in diabetics. Our findings are evidence that abnormally high plasma prorenin levels in diabetics are not an immediate consequence of altered glucose metabolism. This abnormality is related to the development of microvascular disease in the eye and kidney and is at least in part due to decreased clearance of prorenin from the circulation, increased production from extrarenal sources, or both.
Article
Binding and internalization of [125I]angiotensin II (AII) were studied by morphological and biochemical methods in rats in vivo. Light microscope radioautography demonstrated that [125I]AII binds specifically to adrenal zona glomerulosa (ZG) cells. Ultrastructural radioautographic analysis revealed that [125I]AII binds to the cell surface, clusters in coated pits, is internalized in coated vesicles, and is transported by receptosomes to lysosomes in less than 20 min. Biochemical analysis revealed that as much as 40% of the adrenal radioactive uptake behaves as native [125I]AII as shown by electrophoresis, immunoprecipitation and radioligand binding studies. These results indicate that the effects of AII on the secretion of aldosterone by ZG cells are mediated by cell surface phenomena and not by binding to intracellular organelles involved in steroidogenesis. They also indicate that the half-life of AII bound to receptors and internalized seems to be much longer (min) than in the systemic circulation (sec).
Article
Plasma renin exists in an active form or as an inactive zymogen that resembles a prorenin present in homogenates of human kidneys. We examined the relation of diabetes and its microvascular complications with the level of plasma inactive renin activated by dialysis to pH 3.3. Plasma inactive renin was measured in 235 diabetic patients and 90 nondiabetic controls. In the controls, the level of plasma inactive renin increased slightly with age but was never above 50 ng per milliliter per hour. In young diabetic patients studied within three years of the onset of diabetes the concentration of inactive renin was normal, and in some older diabetics without complications it remained within the age-adjusted normal range for many years. However, in patients with retinopathy or albuminuria, plasma inactive renin was above the normal range with few exceptions, reaching levels 50 to 200 per cent above the upper limits of normal in patients with nephropathy. The frequency of neuropathy was also significantly higher among patients with levels above the normal range. In 37 per cent of the diabetics followed during one to three years of conventional treatment, plasma inactive renin increased significantly, but in another group of diabetics under intensive treatment, the level rose in only 7 per cent and fell in 43 per cent. We conclude that there is a close association between a high level of plasma inactive renin and the presence of microvascular complications, and that the level of inactive renin can be modified by intensive treatment of diabetes.
Article
The purpose of this study was to assess the inhibitory effect of TCV-116, an orally active angiotensin II (Ang II) antagonist, on the pressor action of exogenous Ang II and to determine the compensatory rise in plasma renin activity and Ang II levels. Twenty-three male volunteers were treated for 8 days in a double-blind fashion with either placebo or TCV-116 (1, 2, or 4 mg PO daily) and challenged on the first, fourth, and eighth days with repeated bolus injections of Ang II. An additional 4 subjects received 8 mg PO daily in a single-blind fashion. The inhibitory effect on the systolic blood pressure response to Ang II was long lasting and clearly dose related. Six hours after 4 mg TCV-116, the systolic blood pressure response to a given dose of Ang II was reduced to 40 +/- 4% and 35 +/- 8% of baseline value on days 1 and 8, respectively. TCV-116 induced a dose-related increase in plasma renin activity and Ang II levels that was more pronounced on the eighth than on the first day of drug administration. Despite this compensatory mechanism, the relation between the time-integrated systolic blood pressure response to Ang II and the time-integrated CV-11974 levels, the active metabolite of TCV-116, was not different between days 1 and 8. In conclusion, TCV-116 appears to be a well-tolerated, orally active, potent, and long-lasting antagonist of Ang II in men.
Article
Some proteases possess a membrane receptor that focalizes their proteolytic activity on the cell surface and may mediate a proliferative effect, such as urokinase on glomerular epithelial cells. Since some hypertensive states are associated with high concentrations of renin and proliferation of arteriolar smooth muscle cells, we asked whether renin, an aspartyl-protease, would bind to mesangial cells that are smooth-muscle derived cells, which would induce their proliferation. The binding of 125I labeled recombinant human renin (125I-R) was studied on human primary mesangial cells and mesangial cells immortalized by transfection with SV40-T antigen. At 37 degrees C, the binding of 125I-R was time dependent and reached a plateau after two hours. 125I-R was found to bind in a saturable and specific manner with a Kd = 0.4 nM and 1 nM and 8,000 and 2,000 binding sites/cell, for primary and immortalized cells, respectively. When binding experiments were performed in the presence RO 42-5892, a synthetic inhibitor of renin, RO 42-5892 could inhibit the specific binding of labeled renin only at concentrations 1,000 times superior to the IC 50, indicating that the renin-mesangial receptor interaction did not depend on the active site of renin. Analysis by SDS-PAGE and autoradiography of cross-linking experiments of 125I-R bound to a membrane preparation showed a band of approximately 110 to 120 kDa, suggesting a Mr of 70 to 80 kDa for the renin receptor. Incubation of mesangial cells with 100 nM renin for 24 hours provoked a 100% increase of 3H thymidine incorporation that was not accompanied by an increase of the cell number, even after a seven day period of incubation. However, the incubation of mesangial cells with renin for 24 hours induced a significant increase (170% of control, P = 0.04) of plasminogen activator inhibitor-1 (PAI1) antigen in the conditioned medium. In conclusion, we have shown that human mesangial cells in culture express a specific receptor for renin, and that the binding of renin increases 3H thymidine incorporation independently of renin enzymatic activity. The absence of cell proliferation, the increase of 3H thymidine incorporation and the increase of PAI1 antigen suggest that the binding of renin can induce mesangial cell activation, which is reflected by a change in the fibrinolytic capacity of the cells. The role of this receptor remains to be determined in nephropathies and hypertensive states associated with high plasma/tissue renin concentrations, hypertrophy of mesangial or smooth muscle cells and extracellular matrix remodeling.
Article
Angiotensin II (Ang II) is internalized by various cell types via receptor-mediated endocytosis. Little is known about the kinetics of this process in the whole animal and about the half-life of intact Ang II after its internalization. We measured the levels of 125I-Ang II and 125I-Ang I that were reached in various tissues and blood plasma during infusions of these peptides into the left cardiac ventricle of pigs. Steady-state concentrations of 125I-Ang II in skeletal muscle, heart, kidney, and adrenal were 8% to 41%, 64% to 150%, 340% to 550%, and 680% to 2100%, respectively, of the 125I-Ang II concentration in arterial blood plasma (ranges of six experiments). The tissue concentrations of 125I-Ang I were less than 5% of the arterial plasma concentrations. 125I-Ang II accumulation seen in heart, kidney, and adrenal was almost completely blocked by a specific Ang II type 1 (AT1) receptor antagonist. Steady-state concentrations of 125I-Ang II were reached within 30 to 60 minutes in the tissues and within 5 minutes in blood plasma. The in vivo half-life of intact 125I-Ang II in heart, kidney, and adrenal was approximately 15 minutes, compared with 0.5 minute in the circulation. Thus, Ang II, but not Ang I, from the circulation is accumulated by some tissues, and this is mediated by AT1 receptors. The time course of this process and the long half-life of the accumulated Ang II support the contention that this Ang II has been internalized after its binding to the AT1 receptor, so that it is protected from rapid degradation by endothelial peptidases. The results of this study are in agreement with growing evidence of an important physiological role for internalized Ang II.
Article
The binding and internalization of recombinant human renin and prorenin (2500 microU/mL) and the activation of prorenin were studied in neonatal rat cardiac myocytes and fibroblasts cultured in a chemically defined medium. Surface-bound and internalized enzymes were distinguished by the addition of mannose 6-phosphate to the medium, by incubating the cells both at 37 degrees C and 4 degrees C, and by the acid-wash method. Mannose 6-phosphate inhibited the binding of renin and prorenin to the myocyte cell surface in a dose-dependent manner. At 37 degrees C, after incubation at 4 degrees C for 2 hours, 60% to 70% of cell surface-bound renin or prorenin was internalized within 5 minutes. Intracellular prorenin was activated, but extracellular prorenin was not. The half-time of activation at 37 degrees C was 25 minutes. Ammonium chloride and monensin, which interfere with the normal trafficking and recycling of internalized receptors and ligands, inhibited the activation of prorenin. Results obtained with cardiac fibroblasts were comparable to those in the myocytes. This study is the first to show experimental evidence for the internalization and activation of prorenin in extrarenal cells by a mannose 6-phosphate receptor-dependent process. Our findings may have physiological significance in light of recent experimental data indicating that angiotensin I and II are produced at cardiac and other extrarenal tissue sites by the action of renal renin and that intracellular angiotensin II can elicit important physiological responses.