ArticlePDF Available

Anti-necrotic and cardioprotective effects of a cytosolic renin isoform under ischemia-related conditions

Authors:

Abstract and Figures

In the heart, secretory renin promotes hypertrophy, apoptosis, necrosis, fibrosis, and cardiac failure through angiotensin generation from angiotensinogen. Thus, inhibitors of the renin-angiotensin system are among the most potent drugs in the treatment of cardiac failure. Renin transcripts have been identified encoding a renin isoform with unknown targets and unknown functions that are localized to the cytosol and mitochondria. We hypothesize that this isoform, in contrast to secretory renin, exerts cardioprotective effects in an angiotensin-independent manner. Cells overexpressing cytosolic renin were generated by transfection or obtained from CX(exon2-9)renin transgenic rats. Overexpression of cytosolic renin reduced the rate of necrosis in H9c2 cardiomyoblasts and in primary cardiomyocytes after glucose depletion. These effects were not mediated by angiotensin generation since an inhibitor of renin activity did not influence the in vitro effects. siRNA-mediated knockdown of endogenous cytosolic renin increased the rate of necrosis and aggravated the pro-necrotic effects of glucose depletion. Isolated perfused hearts obtained from transgenic rats overexpressing cytosolic renin exhibited a 50 % reduction of infarct size after ischemia-reperfusion injury. Cytosolic renin is essential for survival, both under basal conditions and during glucose starvation. The protective effects are angiotensin-independent and contrary to the known actions of secretory renin. A cytosolic isoform of renin with unknown functions is expressed in the heart. Cytosolic renin diminishes ischemia induced damage to the heart. The protective effects of cytosolic renin contradict the known function of secretory renin. The effects of cytosolic renin are not mediated via angiotensin generation. Renin-binding protein is a potential target for cytosolic renin.
Content may be subject to copyright.
ORIGINAL ARTICLE
Anti-necrotic and cardioprotective effects of a cytosolic renin
isoform under ischemia-related conditions
Heike Wanka
1
& Doreen Staar
1
& Philipp Lutze
1
& Barbara Peters
1
&
Johanna Hildebrandt
1
& Tim Beck
1
& Inga Bäumgen
1
& Alexander Albers
1
&
Thomas Krieg
2,3
& Katrin Zimmermann
3
& Jaroslaw Sczodrok
1
& Simon Schäfer
1
&
Sigrid Hoffmann
4
& Jörg Peters
1
Received: 25 March 2015 /Revised: 25 June 2015 /Accepted: 1 July 2015 /Published online: 11 August 2015
#
Springer-Verlag Berlin Heidelberg 2015
Abstract
In the heart, secretory renin promotes hypertrophy, apoptosis,
necrosis, fibrosis, and cardiac failure through angiotensin gen-
eration from angiotensinogen. Thus, inhibitors of the renin-
angiotensin system are among the most potent drugs in the
treatment of cardiac failure. Renin transcripts have been iden-
tified encoding a renin isoform with unknown targets and
unknown functions that are localized to the cytosol and mito-
chondria. We hypothesize that this isoform, in contrast to se-
cretory renin, exerts cardioprotective effects in an angiotensin-
independent manner. Cells overexpressing cytosolic renin
were generated by transfection or obtained from CX(exon2-
9)renin transgenic rats. Overexpression of cytosolic renin re-
duced the rate of necrosis in H9c2 cardiomyoblasts and in
primary cardiomyocytes after glucose depletion. These effects
were not mediated by angiotensin generation since an inhibi-
tor of renin activity did not influence the in vitro effects.
siRNA-mediated knockdown of endogenous cytosolic renin
increased the rate of necrosis and aggravated the pro-necrotic
effects of glucose depletion. Isolated perfused hearts obtained
from transgenic rats overexpressing cytosolic renin exhibited
a 50 % reduction of infarct size after ischemia-reperfusion
injury. Cytosolic renin is essential for survival, both under
basal conditions and during glucose starvation. The protective
effects are angiotensin-independent and contrary to the known
actions of secretory renin.
Key messages
& A cytosolic isoform of renin with unknown functions is
expressed in the heart.
& Cytosolic renin diminishes ischemia induced damage to
the heart.
& The protective effects of cytosolic renin contradict the
known function of secretory renin.
& The effects of cytosolic renin are not mediated via angio-
tensin generation.
& Renin-binding protein is a potential target for cytosolic
renin.
Keywords Cytosolic renin
.
Ischemia
.
Cardioprotection
.
Glucose depletion
.
Necrosis
Abbreviations
ANG Angiotensin
E(19) Exon(19)renin, secretory
E(1A9) Exon(1A9)renin, cytosolic
E(29) Exon(29)renin, cytosolic
I/R Ischemia/reperfusion
TG+ Transgenic rats overexpressing cytosolic renin
TG- Non-transgenic rats
cAMP Cyclic adenosine monophosphate
Electronic supplementary material The online version of this article
(doi:10.1007/s00109-015-1321-z) contains supplementary material,
which is available to authorized users.
* Heike Wanka
wanka@uni-greifswald.de
1
Department of Physiology, University Medicine of Greifswald,
Greifswalder Str. 11C, D-17495 Karlsburg, Germany
2
Department of Medicine, University of Cambridge, Addenbrookes
Hospital, Cambridge CB2 0QQ, UK
3
Internal Medicine, University Medicine Greifswald,
D-17489 Greifswald, Germany
4
Medical Research Center, Medical Faculty Mannheim, University of
Heidelberg, D-68135 Mannheim, Germany
J Mol Med (2016) 94:6169
DOI 10.1007/s00109-015-1321-z
YWHAZ Tyrosine 3 monooxygenase/tryptophan 5
monooxygenase activation protein, zeta
polypeptide
RLU Relative light units
RnBP Renin-binding protein
RAS Renin-angiotensin system
Introduction
Renin is a secretory glycoprotein that, after secretion into the
extracellular space, generates angiotensin (ANG) I from its
only known substrate, angiotensinogen [1]. ANG I is further
cleaved to ANG II, the effector peptide of the renin-
angiotensin system (RAS) that increases oxidative stress and
exerts pro-inflammatory effects [2, 3]. In the heart, ANG II
promotes hypertrophy, apoptosis, necrosis, fibrosis, myocar-
dial remodeling, and hence, cardiac failure [4, 5]. Correspond-
ingly, inhibitors of the RAS are among the most potent drugs
in the treatment of hypertension and cardiac failure, markedly
increasing the life span of patients [6].
In addition to the classical transcript [exon(19)], alterna-
tive renin transcripts have been identified in several species
incl uding humans [exon(2-9 ), exon(1A-9), renin-c] [79].
The transcript for secretory renin is mainly expressed in the
kidney. The alternative transcripts encode for a non-secretory
cytosolic form of renin and are expressed in extrarenal tissues
[10]. The targeting of proteins to the secretory pathway re-
quires the co-translational transport to the endoplasmic retic-
ulum (ER). In rats, the renin gene consists of nine exons with
the corresponding ER signal sequence, the so-called pre-frag-
ment, being encoded by exon 1. Because the alternative tran-
scripts lack exon 1, the translated renin protein remains in the
cytosol or is taken up by the mitochondria [8]. In humans, the
alternative renin transcript directly starts with exon 2 [9]. In
rats, exon 2 is preceded by a short non-coding sequence of
about 80 base pairs derived from intron A forming the
exon(1A9)renin transcript. The exon(1A9)renin and the ex-
on(29)renin transcripts are translated into a truncated
prorenin starting at the first in-frame UTG in exon 2. This
protein lacks the pre-fragment and the first 15 amino acids
of the conventional prorenin but still exerts renin activity
[79].
The biological functions of cytosolic renin are currently
unknown. In the rat heart, the expression of cytosolic renin
but not of secretory renin was increased after myocardial in-
farction, and first vague hints suggested that cells overexpress-
ing cytosolic renin may be more stable against necrotic cell
death [10, 11]. In this context, it is known that the heart pos-
sesses the ability to adapt to stress by actively enhancing its
resistance to injury. This molecular plasticity is illustrated by
the phenomenon of ischemic preconditioning where the toler-
ance of the heart to an ischemic event is increased by
influencing different signaling pathways [12, 13]. Similar ef-
fects described as part of a genetic reprogramming to fetal
gene profile are also detectable in failing hearts [14, 15]. We
hypothesized that cytosolic renin may play a role in the re-
sponse to energy depletion and that it may reduce the severity
of cardiac damage under ischemic conditions. In this study, we
show that cytosolic renin decreases the rate of necrosis, par-
ticularly under conditions of glucose depletion and attenuates
the infarct size after coronary ligation in isolated beating
hearts.
Methods
Overexpression, downregulation and inhibition of renin
Hearts from CX-exon(2-9)renin transgenic rats (TG+) over-
expressing cytosolic renin under the control of the CX-CMV
promoter and hearts from non-transgenic control rats (TG-)
were used to prepare primary cardiomyocytes [16]. H9c2
cardiomyoblasts (ATCC, CRL-1446) were transfected with
the pIRES1neo vector containing exon(2-9)renin DNA as pre-
viously described [11]. Downregulation of renin in H9c2 cells
was accomplished with the RNA interference method using
80 nmol/L siGENOME SMART pool siRNA to renin (Ther-
mo Scientific Dharmacon) and polyethyleneimine solution
according to the manufacturers instructions. Renin transcript
abundance was quantified using qRT-PCR and Western blot
analyses. The renin inhibitor CH732 (10
9
to 10
6
mol/L) was
used to block any possible generation of ANG I by exogenous
renin [1.8 μg ANG I/(ml×h)]. After incubation of H9c2 cells
with 10
6
mol/L CH732 for 24 h, trypsinated cells were
sonificated and separated into a soluble and a membrane frac-
tion by centrifugation at 20,000×g. The pellet and the soluble
fractions potentially containing CH732 were then used to in-
hibit renin enzyme activity in terms of ANG I generation [17]
as determined with renin standard preparations (see
Supplement Material 1). For functional analysis, CH732 was
added to H9c2 cells for 24 h in a concentration of 10
6
mol/L
followed by the detection of the necrosis rate.
Analysis of metabolic parameters and necrosis in cardiac
cells
Cardiomyoblasts (H9c2, ATCC) and primary cardiomyocytes
were cultured at 37 °C in a humidified atmosphere with 5 %
CO
2
. Plated cells were subjected to control conditions or glu-
cose deprivation for 24 or time dependent for 24, 72, and
120 h, respectively. Consequences of glucose deprivation on
necrosis rate were analyzed by the Cytotoxicity Detection Kit
(LDH) (Roche Applied Science, Germany) as previously de-
scribed [11]. ATP content, glucose consumption and lactate
62 J Mol Med (2016) 94:6169
accumulation into the medium were detected as described in
the expanded methods online supplement.
Analysis of renin expression and renin/prorenin activities
After treatment of cardiac cells, renin expression was deter-
mined by quantitative realtime PCR (Rotor-Gene SYBR
Green PCR Kit) and Western blot analyses. For quantification
of specific renin transcript isoforms, the following primers
were used: renin reverse primer: position 722-743 of the renin
cDNA and the following forward primers for specific tran-
scripts: exon(1A9)renin: position 3996-4019 of intron A of
the renin gene, exon(19)renin: position 211-233 of the renin
cDNA, exon(29)renin: position 421442 of the renin cDNA.
The threshold cycle number (CT) in combination with the
2
delta CT
method was normalized against YWHAZ expression
and compared to the control.
To detect renin protein, we used a primary rabbit anti-renin
antibody (Bioss Inc, Woburn, MA, USA) in combination with
a horseradish peroxidase (HRP)-conjugated secondary anti-
rabbit antibody (CellSignaling, Merck-Millipore, Billerica,
MA, USA). The proteins were visualized by the enhanced
chemiluminescence method (BioRad Laboratories, Munich,
Germany) using an image capture system (Chemidoc XRS,
BioRad Laboratories, Munich, Germany).
Renin activity was determined as follows. Extracts from
H9c2 cell lines were prepared by ultrasonification. Mem-
branes were pelleted by centrifugation at 20,000× g and
discarded. (Pro)renin activities of the supernatants were deter-
mined by measuring the capability of the extracts to generate
ANG I from excess renin substrate followed by an ANG I
radioimmunoassay as previously described [11, 16]. To fur-
ther test the specificity of the results, aliquots of extracts were
incubated with and without the addition of the rat renin inhib-
itor CH732 [17]. The inhibitor completely abolished the gen-
eration of ANGI in all H9c2 cell lines.
Langendorff experiment
TG+ and TG rats were deeply anesthetized with thiopental
solution containing heparin to remove their hearts for
Langendorff experiments. Cardiac ischemia was induced by
placing a ligature around the left anterior descending artery for
30 min followed by 120 min reperfusion. A solution of mi-
crospheres was injected retrograde though the aortic root to
demarcate the risk zone. Transverse slices of the hearts were
stained with triphenyltetrazolium chloride (TTC) to assess the
non-infarcted myocardium of the risk zone [18].
Additionally, detailed material and methods are provided in
the online supplement.
Results
Upregulation of cytosolic renin
During periods of ischemia, cells are deprived of extra-
cellular glucose. Here, we demonstrate that in H 9c2
cardiomyoblasts, glucose depletion for 24 h upregulated
the expression of cytosolic exon(1A9)renin but not of
secretory exon(19)r enin at the transcript and protein
levels (Fig. 1ac). Although renin expression clearly
increased at mRNA (Fig. 1a) and protein levels
(Fig. 1b, c), the renin activity even decreased to unde-
tectable levels (not shown), likely due to an inhibitory
effect of cytosolic renin-binding protein (RnBP).
Fig. 1 Cytosolic renin expression is increased in cardiac H9c2 cells
under glucose deprivation. Cells were cultured in medium containing
25 mM glucose (white columns) or exposed to glucose deprivation for
24 h (gray columns). a Effect of 24 h glucose deprivation on transcript
levels of cytosolic exon(1A-9)renin and secretory exon(19)renin as
detected by RT PCR. Expression of renin transcripts was normalized to
YWHAZ as housekeeping gene. b Western blot demonstrates the effect
of glucose deprivation on renin protein expression. Bands corresponding
to cytosolic prorenin and renin are estimated at 30, 36, and 41 kDa,
respectively (arrows). c Renin protein normalized to total protein
content. Values repres ent means±s.e.m., n =8 experiments each;
*p<0.05, **p<0.01
J Mol Med (2016) 94:6169 63
Cytosolic renin protects cardiac cells from necrosis
To determine the functional relevance of cytosolic renin,
we evaluated whether or not cytosolic renin can protect
cardiac H9c2 cells from necrosis during glucose deple-
tion. To this end, we generated a number of cell lines
overexpressing cytosolic renin at various degrees rang-
ing from 3- to 2000-fold (confirmed by qRTPCR as
well as by We stern blot, not shown). We then chose
such cells overexpressing renin by about 5- to 10-fold,
which compares well with the amount of overexpression
after glucose depletion (Fig. 1). Renin mRNA was in-
creased by about 8-fold (Fig. 2a). Nevertheless, renin as
determined by its capacity to generate ANGI from ex-
cess substrate was not increased (Fig. 1c). This may be
attributed to the inhibitory effect of RnBP present in
those cells. However, after trypsin activation, the over-
expression was also obvious at the protein/enzymatic
level (Fig. 1b).
In the presence of glucose, cells overexpressing cytosolic
renin by about 5- to 10-fold (Fig. 2ac) exhibited low necrosis
rates similar to H9c2 and pIRES vector-transfected cells.
Twenty-four hours of glucose depletion markedly increased
necrosis rates in control cells. In contrast, in cells overexpress-
ing cytosolic renin, the increase of necrosis rate in response to
glucose depletion was abolished (Fig. 2d).
This anti-necrotic effect was further validated in primary
cardiomyocytes derived from transgenic rats overexpressing
cytosolic renin in the heart [16]. Primary cardiomyocytes of
TG+ rats were also protected from necrotic cell death under
conditions of glucose deprivation, however with a delay com-
pared with the H9c2 cell lines (Fig. 3). Anti-necrotic effects
were detectable after 72 and 120 h of glucose deprivation.
Vice versa, knockdown of cytosolic renin expression by
siRNA should diminish survival of cardiomyoblasts. In our
experiments, we used a 48 h knockdown because the efficien-
cy of knockdown decreased time dependently (Fig. 4a). After
knockdown, necrosis rates of H9c2 cells increased already
under basal conditions and the increase after glucose depletion
was still prominent (Fig. 4b).
Cytosolic renin acts ANG-independently
To clarify whether the anti-necrotic effects of cytosolic renin
were ANG-mediated, we applied the rat-specific renin inhib-
itor CH732. CH732 inhibited renin activity dose dependently
and almost completely at a conc entr ation of 10
6
mol/L
(Fig. 5a). Furthermore, CH732 inhibitory activity was ob-
served in both the soluble and the membrane fraction s of
CH732-pretreated H9c2 cells indicating the uptake of
CH732 into the cells (Fig. 5b). Although CH732 clearly
inhibited ANG generation and was taken up by the cells
(Fig. 5a, b), CH732 did not block the protective anti-necrotic
effects of cytosolic renin overexpression (Fig. 5c).
Cytosolic renin changes metabolic state
Under basal conditions, cells overexpressing cytosolic renin
differed from vector-transfected pIRES cells by a modified
metabolic state as evidenced by a reduced ATP content (788
±59×10
3
vs. 991±18×10
3
RLU; n=9; p<0.01), a higher glu-
cose consumption (10.80±0.43 vs. 6.08±0.66 mM/10
6
cells/
24 h; n=8; p<0.001), and an increased accumulation of lactate
in the medium (9.28±1.74 vs. 5.14±0.49 mM/10
6
cells/24 h;
n=8; p<0.05). Furthermore, glucose deprivation for 24 h de-
creased ATP content in vector-transfected control cells but not
Fig. 2 Cytosolic renin protects H9c2 cells from necrotic cell death. a
Transcript levels of cytosolic exon(29)reninnormalizedtoYWHAZas
detected by RT PCR. b, c Enzymatic inactive (prorenin) and active renin
of H9c2 cell lines. In extracts from exon(29)renin overexpressing H9c2
cells, the prorenin level (a, dark column) was significantly increased
compared to H9c2 and pIRES control cells (each n=6). There were no
differences in renin activity (b). d Effect of cytosolic renin overexpression
on LDH release. H9c2 cells (white column), pIRES control cells (gray
columns) and H9c2 cells overexpressing cytosolic exon(2-9)renin (dark
columns) were maintained in medium containing 25 mM glucose (+) or
exposed to glucose depletion (-) for 24 h (each n=9). The values shown
are mean±s.e.m. Statistical analysis was performed by one way ANOVA
with *p<0.05 and ***p<0.001 as indicated and
###
p<0.001 effect
between exon(2-9) and pIRES control cells grown under glucose
depletion
64 J Mol Med (2016) 94:6169
in cells overexpressing cytosolic renin (controls: from 991±
18 to 855±21×10
3
vs. E(29) cells: from 788±59 to 841±
55×10
3
RLU; n=9; p<0.01).
Protective effects of cytosolic renin in isolated perfused
hearts
We finally applied an ex vivo model of myocardial infarction
to further investigate the protective effects of cytosolic renin
under conditions of ischemia reperfusion (I/R) induced by
coronary ligation. For these experiments, we used two inde-
pendently generated transgenic rat lines and two non-
transgenic control lines. In SpragueDawley and Wistar rat
hearts, the infarct size relative to the area at risk was about
40 %. In both strains of TG+ rats, infarct size was reduced to
about 20 % of the risk area, representing a 50 % reduction of
the infarct size compared to controls (Fig. 6).
Discussion
There is no doubt that renin-angiotensin systems reside locally
within the heart [19, 20]. Even the existence of intracellular
acting RAS has been postulated, but this is presently subject of
controversial discussions [21]. However, it is difficult to un-
derstand how secretory proteins such as renin and
angiotensinogen can be located in the cytosol, in mitochon-
dria, or in nuclei since these proteins are inevitably packed
into secretory vesicles for externalization or internalized via
vesicle-mediated uptake . Meanwhile, for renin but not for
angiotensinogen, alternative transcripts have been discovered
coding for a renin isoform that is translated at free ribosomes
[79]. This renin isoform is located in the cytosol and can be
imported into mitochondria [8]. Here, we report that this cy-
tosolic renin protects cardiac cells from necrosis and reduces
infarct sizes in isolated perfused hearts. We also demonstrate
that the anti-necrotic effect of cytosolic renin is not mediated
by cleaving ANG I from angiotensinogen and suggest the
renin-binding protein (RnBP) as a possible target for cytosolic
renin. The fact that renin expression increased by glucose
starvation (mRNA and protein levels) whereas renin activity
remained unchanged supports the hypothesis that in the cyto-
sol renin activity is blocked, and RnBP is a likely candidate
[22] since RnBP forms heterodimers with renin particularly
under energy depletion [23].
During ischemia, the expression of cytosolic renin is up-
regulated in vivo [10]. Here , we demonstrate that glucose
starvation as one aspect of ischemia also upregulates the en-
dogenous expression of cytosolic renin but not of secretory
renin, indicating that especially the cytosolic renin isoform is
part of an adaptive response to starvation. Applying different
models of overexpression and downregulation of cytosolic
renin, we come to the conclusion that endogenous cytosolic
Fig. 3 Anti-necrotic effect of cytosolic renin in primary cardiomyocytes.
Cardiac cells overexpressing cytosolic renin obtained from TG 294 and
TG 307 (n=7 each) (gray columns) and cardiac cells from control TG
rats ( n=7) (white columns) were maintained in medium containing
25 mM glucose or exposed to glucose depletion (0 mM) for 24 h (a),
72 h (b), or 120 h (c), respectively. Cardiomyocytes of TG+ rats were
protected from necrotic cell death caused by glucose deprivation after 72
and 120 h. Data are presented as means±s.e.m.; *p<0.05 vs. TG;
**p<0.01 vs. TG;
#
p<0.05 vs. basal conditions;
##
p<0.01 vs. basal
conditions
J Mol Med (2016) 94:6169 65
renin protects cardiac cells under ischemia and ischemia-
related conditions. For the present studies, we used CX-ex-
on(29)renin transgenic rats overexpressing cytosolic renin
about 5 -fold in the h eart and accordingly exon(29)renin
transfected H9c2 cardiomyoblasts showing a 5- to 10-fold
overexpression of cytosolic renin. We previously demonstrat-
ed already that overexpression of cytosolic renin in cells as
well as in rats resulted in an increase of renin particularly in
the cytosolic or mitochondrial fractions [11, 16]. The degree
of overexpression of cytosolic renin corresponds well with
that seen after myocardial infarction in vivo [10, 24]. To ex-
clude or at least minimize genetic background effects, we
chose two independently generated transgenic rat strains, line
294 and 307, as well as two different control strains (Wistar,
SpragueDawley rats from different providers) for the exper-
iments. The rationale was that all rats without overexpression
of cytosolic renin should be susceptible to ischemia indepen-
dently of further genetic differences, whereas all rats with
overexpression of cytosolic renin were protected, again inde-
pendently of further genetic differences. In transgenic rats, the
Fig. 4 Renin knockdown increased necrotic cell death in H9c2 control
cells. a Time dependent efficiency of siRNA-mediated downregulation of
exon(1A-9)renin transcript validated by qRT-PCR. Renin expression was
normalized to YWHAZ as housekeeping gene and related to untreated
scramble controls. b Effect of 48 h downregulation of endogeneous
cytosolic renin on LDH release of control and glucose-deprived H9c2
cells. Values represent means±s.e.m. of n=8 experiments. Statistical
analysis was performed by one way (a) and two-way ANOVA (b)with
*p< 0.05; ***p <0.001 as indicated, and
§§
p<0.01 effect of siRNA-
mediated downregulation vs. untreated cells grown under the same
conditions
Fig. 5 Inhibition of renin activity by CH732 did not prevent anti-necrotic
effects of cytosolic renin. a Dose-response curve of CH732 on the
efficacy to block ANG I generation as determined by the
radioimmunoassay. Complete inhibition of renin activity was reached at
a CH732 concentration of 10
-6
mol/L (n=6). b Inhibitory potential of
H9c2 cell fractions pretreated with 10
-6
mol/L CH732 for 24 h. The
data confirm that the renin inhibitor wa s taken up by the cells and
appeared in the cytosolic fraction where it effectively reduced
exogenous renin activity (n=6). c Anti-necrotic, protective effects of
cytosolic renin under glucose deprivation were not impaired by CH732
pretreatment. Experiments wereperformedwithpIREScells(gray
columns) and E(2-9)renin overexpressing cells (dark columns) for 24 h
without and with a concentration of 10
-6
mol/L (n=9). Values represent
means±s.e.m. Statistical analysis was performed by one way (a, b)and
two-way ANOVA (c)with*p>0.05; **p<0.01; ***p< 0.001 as indicated
and ###p<0.001 effect of cytosolic renin vs. pIRES control cells grown
under the same conditions
66 J Mol Med (2016) 94:6169
levels of circulating renin and prorenin were not different from
controls. Rats overexpressing cytosolic renin showed no signs
of inflammation, fibrosis, hypertrophy, or cardiac failure, ex-
cluding harmful effects of cytosolic renin in vivo [16].
In the presence of glucose, H9c2 cells overexpressing cy-
tosolic renin exhibited low necrosis rates similar to vector-
transfected control cells, confirming the lack of toxicity of
cytosolic renin under basal conditions in vitro. During glucose
starvation, overexpression of cytosolic renin prevented the
rise in necrosis rates. O n the other hand, t he siRNA-
mediated knockdown of renin expression in H9c2 cells was
already harmful under basal conditions by increasing the ne-
crosis rate. These data confirm the specificity of our findings.
Compared to H9c2 cells, primary cardiomyocytes showed a
markedly higher basal necrosis rate which may be due to the
isolation procedure. However, they seem to be more resistant
to glucose depletion in general because starvation did not
enhance necrosis rate. Overexpression of cytosolic renin in
cardiomyocytes obtained from transgenic rats resulted in a
marked reduction of necrosis rates compared to non-
transgenic primary cardiomyocytes, demonstrating the
cardioprotective potential of cytosolic renin. The data support
the hypothesis that endogenous cytosolic renin substantially
enhances the survival of cardiac cells both under basal and
especially under glucose-deprived conditions in vitro. The
cardioprotective effects of cytosolic renin may be particularly
beneficial during transient ischemic periods in patients with
coronary heart disease.
To clarify the underlying protective mechanisms, especial-
ly to check for potential intracrine ANG effects, we used the
rat renin inhibitor CH732 [17]. Although the inhibitor clearly
inhibited ANG I generation in vitro and was taken up into the
cells, CH732 did not block the protective effects of cytosolic
renin in glucose-deprived cells. These data demonstrate that
the cardioprotective effects of cytosolic renin are ANG-inde-
pendent. In contrast, the glucose depletion-induced rise in
necrosis rate was higher in CH732-pretreated pIRES cells than
in untreated pIRES control cells. In this context, the existence
of an intracellular RAS with intra-cytoplasmatic ANG II ac-
tions has long been postulated (for review, see [21]). There is
evidence for the presence of renin, angiotensin-converting en-
zyme, ANGs, and ANG receptors in the cytosol, nuclei, and
mitochondria. A complete mitochondrial RAS has been re-
ported in various cell types including mouse cardiac myocytes
[25]. Intracellular application of renin decreased junctional
conductance between cardiac cells, which was potentiated
by concomitantly dialyzed angiotensinogen. Also, ANG II
injected into the cytoplasm increased cytosolic calcium, which
was blocked by concomitant injection of an AT1 receptor
blocker [26, 27]. Furthermore, in isolated nuclei, ANG II stim-
ulated the de novo synthesis of RNA via an ANG receptor-
dependent mechanism [28]. Overexpression of
angiotensinogen increased the mitogenic index, which was
blocked by the AT1 receptor blocker losartan and co-
expression of ANG II together with the AT1 receptor en-
hanced cell proliferation and increased the activity of cAMP
response element-binding protein (CREB) [29, 30]. It is im-
portant to note that in these studies, ANG II was given intra-
cellular by microinjections or by dialysis or produced from
engineered constructs encoding a non-secretory
angiotensinogen variant or an ANG II fusion protein. It still
remains unclear how ANG can be generated within the cyto-
plasm and how it can reach the mitochondria or the nucleus,
assuming that angiotensinogen is a secretory protein and
should not be localized in the cytosol. Our data neither support
nor exclude the idea of intracellular actions of ANGs; however ,
the fact that the renin inhibitor CH732 did not abolish the
protective effects of cytosolic renin demonstrates that these
effects are not mediated by ANGs. The effect of CH732 on
control cells, however, supports the view that additionally
intracellular ANG may have so far unknown effects.
In diabetic rats, a hyperglycemia-associated upregulation
of the RAS contributes to an increase of intracellular produced
ANG II that correlates with cardiomyocyte apoptosis, en-
hanced oxidative stress, and cardiac fibrosis [31]. The group
also demonstrated that the renin inhibitor aliskiren prevented
intracellular ANG II synthesis and reduced harmful effects
more efficiently than other RAS blockers in cardiac fibroblast
under glucose load [32]. In their study, an increase of renin
protein levels was observed in response to glucose load
in vitro. Whether or not this was the result of increased
Fig. 6 Hearts of cytosolic renin transgenic rats are protected against
ischemia-reperfusion injury ex vivo. Effect of 30 min ischemia
followed by 120 min reperfusion on infarct size in isolated perfused
Langendorff hearts of two control rat lines (SD; W) and two
independently generated transgenic rat lines overexpressing cytosolic
renin in the heart (line 294; 307). Infarct size was reduced in transgenic
rats (line 294: n=8 and line 307: n=9) compared to control rats (each n=
10). Data are presented as means±s.e.m.; *p<0.05
J Mol Med (2016) 94:6169 67
expression of secretory or cytosolic renin still remains to be
investigated. Since the increased renin protein levels were
accompanied by increased intracellula r angiotensin levels
and harmful effects as shown by Singh et al., it is likely that
under glucose load in this experiment, the expression of
secretory renin was increased rather than of cytosolic renin.
The intracellular interaction partners for cytosolic renin
still need to be identified. One possible target is the cyto-
solic RnBP that has already been shown to interact with
renin in vitro particularly under conditions with depletion
of high-energy nucleotides [23, 33]. Such conditions may
prevail during ischemia. RnBP ac ts as an epimerase,
interconverting N-acetyl-D-glucosamine and N-acetyl-D-
mannosamine. This enzymatic activity of RnBP is inhibited
by renin [34]. By regulating the levels of N-acetyl-D-glu-
cosamine, RnBP may affect the glycosylation and
sialylation of many proteins. As a first hint, downregulation
of cytosolic renin increased the level of free sialic acids
released into the medium and a mild siRNA-mediated
downregulation of RnBP had similar protective effects as
overexpression of cytosolic renin (data not shown). Thus,
cytosolic renin may be an endogenous inhibitor of Nace-
tylDglucosamine epimerase activity. However, other
mechanisms of action also need to be considered.
An important principle of cardioprotection against ische-
mia is the adequate provision of ATP [35]. Opie first described
the so-called glucose hypothesis according to which an en-
hanced uptake and metabolism of glucose delays cellular
damage [36]. In agreement with this hypothesis, our cells
overexpressing cytosolic renin exhibited an enhanced basal
exogenous glucose uptake and increased lactate accumulation
in the medium compared to H9c2 and pIRES control cells
indicating a metabolic alteration. This alteration is not associ-
ated with an anaerobic exploitation of glucose because the
ratio of lactate release to glucose uptake did not differ between
the cell lines. Instead, this effect may be achieved by a raised
generation or storage of glycogen since glucose was the sole
fuel in medium of the cell culture experiments. This assump-
tion is supported by our results that following glucose starva-
tion cells overexpressing cytosolic renin exhibited an en-
hanced preservation of the ATP content. In conclusion, these
data suggest that cells overexpressing cytosolic renin may be
better adapted to fight fuel deficiencies than control cells pro-
viding them with stronger protection against acute ischemia-
mediated cell death.
We finally applied an ex vivo model of myocardial infarc-
tion to investigate the effects of cytosolic renin under condi-
tions of I/R. For these experiments, we used two independent-
ly generated transgenic rat lines and two different non-
transgenic control lines in an attempt to minimize artifacts
due to transgene-induced insertional mutagenesis or due to
differences in the genomic background. The remarkable re-
duction in infarct size in hearts from transgenic rats compared
to that in hearts from control rats demonstrates that cytosolic
renin protects the heart also from I/R injury. Additional studies
are necessary to determine the mechanisms and signal path-
ways by which cytosolic renin elicit cardioprotection. Several
in vivo studies have shown that aliskiren, a clinically used oral
renin inhibitor, is able to abrogate the detrimental cardiac ef-
fects of I/R in rats independent of blood pressure lowering [37,
38]. Such studies underline the harmful effects of angiotensin
generation and hence the potential benefit of renin inhibitors.
Our data do not contradict these observations since we dem-
onstrate that inhibition of renin activity does not play a role for
the functions of cytosolic renin including those presented in
this study.
In conclusion, cytosolic renin has the potential of becoming
a new therapeutic target for the treatment of cardiac diseases
as it decreases necrotic cell death and reduces infarct size after
I/R. It is quite likely that this new cytosolic renin system is not
cardi o-specific but may also protect other cell types from
death under ischemic conditions.
Acknowledgements The work was supported by a grant from the Ger-
man Research Foundation to J. Peters (PE 366/11-1). B. Sturm and D.
Albrecht performed technical assistance.
Conflict of interest The authors declare that they have no competing
interests.
References
1. Hackenthal E, Paul M, Ganten D, Taugner R (1990) Morphology,
physiology, and molecular biology of renin secretion. Physiol Rev
70:10671116
2. Paul M, Poyan Mehr A, Kreutz R (2006) Physiology of local renin-
angiotensin systems. Physiol Rev 86:747803
3. Unger T (2002) The role of the renin-angiotensin system in the
development of cardiovascular disease. Am J Cardiol 89:3A9A,
discussion 10A
4. Aoki H, Richmond M, Izumo S, Sadoshima J (2000) Specific
role of the extracellular signal-regulated kinase pathway in an-
giotensin II-induced cardiac hypertrophy in vitro. Biochem J
347(Pt 1):275284
5. Mehta PK, Griendling KK (2007) Angiotensin II cell signaling:
physiological and pathological effects in the cardiovascular system.
Am J Physiol Cell Physiol 292:C8297
6. Werner C, Baumhakel M, Teo KK, Schmieder R, Mann J, Unger T,
Yusuf S, Bohm M (2008) RAS blockade with ARB and ACE in-
hibitors: current perspective on rationale and patient selection. Clin
Res Cardiol 97:418431
7. Lee-Kirsch MA, Gaudet F, Cardoso MC, Lindpaintner K (1999)
Distinct renin isoforms generated by tissue-specific transcription
initiation and alternative splicing. Circ Res 84:240246
8. Clausmeyer S, Sturzebecher R, Peters J (1999) An alternative tran-
script of the rat renin gene can result in a truncated prorenin that is
transported into adrenal mitochondria. Circ Res 84:337344
9. Sinn PL, Sigmund CD (2000) Identification of three human renin
mRNA isoforms from alternative tissue-specific transcriptional ini-
tiation. Physiol Genomics 3:2531
68 J Mol Med (2016) 94:6169
10. Clausmeyer S, Reinecke A, Farrenkopf R, Unger T, Peters J (2000)
Tissue-specific expression of a rat renin transcript lacking the cod-
ing sequence for the prefragment and its stimulation by myocardial
infarction. Endocrinology 141:29632970
11. Wanka H, Kessler N, Ellmer J, Endlich N, Peters BS, Clausmeyer S,
Peters J (2009) Cytosolic renin is targeted to mitochondria and
induces apoptosis in H9c2 rat cardiomyoblasts. J Cell Mol Med
13:29262937
12. Heusch G (2013) Cardioprotection: chances and challenges of its
translation to the clinic. Lancet 381:166175
13. Hausenloy DJ (2013) Cardioprotection techniques: precondition-
ing, postconditioning and remote conditioning (basic science).
Curr Pharm Des 19:45444563
14. De Mello WC (2006) Cardiac intracrine renin angiotensin system.
Part of genetic reprogramming? Regul Pept 133:1012
15. Kim SH, Kim HS, Lee MM (2002) Re-expression of fetal troponin
isoforms in the postinfarction failing heart of the rat. Circ J 66:959
964
16. Peters J, Wanka H, Peters B, Hoffmann S (2008) A renin transcript
lacking exon 1 encodes for a non-secretory intracellular renin that
increases aldosterone production in transgenic rats. J Cell Mol Med
12:12291237
17. Peters J, Kranzlin B, Schaeffer S, Zimmer J, Resch S, Bachmann S,
Gretz N, Hackenthal E (1996) Presence of renin within
intramitochondrial dense bodies of the rat adrenal cortex. Am J
Physiol 271:E439450
18. Maas O, Donat U, Frenzel M, Rutz T, Kroemer HK, Felix SB,
Krieg T (2008) Vardenafil protects isolated rat hearts at reperfusion
dependent on GC and PKG. Br J Pharmacol 154:2531
19. Peters J, Clausmeyer S (2002) Intracellular sorting of renin: cell
type specific differences and their consequences. J Mol Cell
Cardiol 34:15611568
20. Peters J (2008) Secretory and cytosolic (pro)renin in kidney, heart,
and adrenal gland. J Mol Med (Berl) 86:711714
21. Peters J (2013) Cytosolic (pro)renin and the matter of intracellular
renin actions. Front Biosci (Schol Ed) 5:198205
22. Takahashi S, Inoue H, Fukui K, Miyake Y (1994) Structure and
function of renin binding protein. Kidney Int 46:15251527
23. Takahashi S, Hori K, Ogasawara H, Hiwatashi K, Sugiyama T
(2006) Effects of nucleotides on the interaction of renin with
GlcNAc 2-epimerase (renin binding protein, RnBP). J Biochem
140:725730
24. Passier RC, Smits JF, Verluyten MJ, Daemen MJ (1996) Expression
and localization of renin and angiotensinogen in rat heart after
myocardial infarction. Am J Physiol 271:H10401048
25. Abadir PM, Foster DB, Crow M, Cooke CA, Rucker JJ, Jain A,
Smith BJ, Burks TN, Cohn RD, Fedarko NS et al (2011)
Identification and characterization of a functional mitochondrial
angiotensin system. Proc Natl Acad Sci U S A 108:1484914854
26. De Mello WC (1998) Intracellular angiotensin II regulates the in-
ward calcium current in cardiac myocytes. Hypertension 32:976
982
27. Haller H, Lindschau C, Erdmann B, Quass P, Luft FC (1996)
Effects of intracellular angiotensin II in vascular smooth muscle
cells. Circ Res 79:765772
28. Tadevosyan A, Maguy A, Villeneuve LR, Babin J, Bonnefoy A,
Allen BG, Nattel S (2010) Nuclear-delimited angiotensin receptor-
mediated signaling regulates cardiomyocyte gene expression. J Biol
Chem 285:2233822349
29. Cook JL, Zhang Z, Re RN (2001) In vitro evidence for an intracel-
lular site of angiotensin action. Circ Res 89:11381146
30. Cook JL, Re R, Alam J, Hart M, Zhang Z (2004) Intracellular
angiotensin II fusion protein alters AT1 receptor fusion protein dis-
tribution and activates CREB. J Mol Cell Cardiol 36:7590
31. Singh VP, Le B, Khode R, Baker KM, Kumar R (2008) Intracellular
angiotensin II production in diabetic rats is correlated with cardio-
myocyte apoptosis, oxidative stress, and cardiac fibrosis. Diabetes
57:32973306
32. Singh VP, Baker KM, Kumar R (2008) Activation of the intracel-
lular renin-angiotensin system in cardiac fibroblasts by high glu-
cose: role in extracellular matrix production. Am J Physiol Heart
Circ Physiol 294:H1675
1684
33
. Inoue H, Fukui K, Takahashi S, Miyake Y (1990) Molecular clon-
ing and sequence analysis of a cDNA encoding a porcine kidney
renin-binding protein. J Biol Chem 265:65566561
34. Takahashi S, Kumagai M, Shindo S, Saito K, Kawamura Y (2000)
Renin inhibits N-acetyl-D-glucosamine 2-epimerase (renin-binding
protein). J Biochem 128:951956
35. Schulz R, Cohen MV, Behrends M, Downey JM, Heusch G (2001)
Signal transduction of ischemic preconditioning. Cardiovasc Res
52:181198
36. Opie LH (1990) Myocardial ischemiametabolic pathways and
implications of increas ed glycolysis. Cardiovasc Drugs Ther
4(Suppl 4):777790
37. Zhang W, Han Y, Meng G, Bai W, Xie L, Lu H, Shao Y, Wei L, Pan
S, Zhou S et al (2014) Direct renin inhibition with aliskiren protects
against myocardial ischemia/reperfusion injury by activating nitric
oxide synthase signaling in spontaneously hypertensive rats. J Am
Heart Assoc 3, e000606
38. Koid SS, Ziogas J, Campbell DJ (2014) Aliskiren reduces myocar-
dial ischemia-reperfusion injury by a bradykinin B2 receptor- and
angiotensin AT2 receptor-mediated mechanism. Hypertension 63:
768773
J Mol Med (2016) 94:6169 69

Supplementary resource (1)

... To generate stable renin-b-overexpressing cell lines, transfected cells were cultured in the presence of G418 sulfate (430 µg/mL, Gibco) to ensure the survival of transfected cells only. (1)(2)(3)(4)(5)(6)(7)(8)(9), including the classical exon 1 as well as the alternative transcripts Ren(1a-9) and Ren (2)(3)(4)(5)(6)(7)(8)(9), where Ren(1a-9) comprises the 5′ UTR located in exon 1a and Ren (2)(3)(4)(5)(6)(7)(8)(9) is missing it. The transcription of Ren (1)(2)(3)(4)(5)(6)(7)(8)(9) starts at AUG in exon 1, while for Ren(1a-9) and Ren (2)(3)(4)(5)(6)(7)(8)(9), the start codon is located in exon 2, resulting in either the classical secretory renin-a or the alternative non-secretory renin-b. ...
... To generate stable renin-b-overexpressing cell lines, transfected cells were cultured in the presence of G418 sulfate (430 µg/mL, Gibco) to ensure the survival of transfected cells only. (1)(2)(3)(4)(5)(6)(7)(8)(9), including the classical exon 1 as well as the alternative transcripts Ren(1a-9) and Ren (2)(3)(4)(5)(6)(7)(8)(9), where Ren(1a-9) comprises the 5′ UTR located in exon 1a and Ren (2)(3)(4)(5)(6)(7)(8)(9) is missing it. The transcription of Ren (1)(2)(3)(4)(5)(6)(7)(8)(9) starts at AUG in exon 1, while for Ren(1a-9) and Ren (2)(3)(4)(5)(6)(7)(8)(9), the start codon is located in exon 2, resulting in either the classical secretory renin-a or the alternative non-secretory renin-b. ...
... To generate stable renin-b-overexpressing cell lines, transfected cells were cultured in the presence of G418 sulfate (430 µg/mL, Gibco) to ensure the survival of transfected cells only. (1)(2)(3)(4)(5)(6)(7)(8)(9), including the classical exon 1 as well as the alternative transcripts Ren(1a-9) and Ren (2)(3)(4)(5)(6)(7)(8)(9), where Ren(1a-9) comprises the 5′ UTR located in exon 1a and Ren (2)(3)(4)(5)(6)(7)(8)(9) is missing it. The transcription of Ren (1)(2)(3)(4)(5)(6)(7)(8)(9) starts at AUG in exon 1, while for Ren(1a-9) and Ren (2)(3)(4)(5)(6)(7)(8)(9), the start codon is located in exon 2, resulting in either the classical secretory renin-a or the alternative non-secretory renin-b. ...
Article
Full-text available
The classical secretory renin-a is known to be involved in angiotensin generation, thereby regulating not only blood pressure, but also promoting oxidative stress as well as apoptotic and necrotic cell death. In contrast, another cytosolic renin isoform named renin-b has been described, exerting protective effects under ischemia-related conditions in H9c2 cardiomyoblasts. Using microarray-based transcriptome analyses, we aimed to identify the signaling pathways involved in mediating cardioprotection in H9c2 cells overexpressing renin-b. By transcriptome profiling, we identified increased gene expression of several genes encoding glycolytic enzymes and glucose transporters, while the transcript levels of TCA-cycle enzymes were decreased. Complementing data from metabolic analyses revealed enhanced glucose consumption and lactate accumulation due to renin-b overexpression. Renin-b overexpression further stimulated AKT/mTOR signaling, where numerous genes involved in this pathway showed altered transcript levels. For AKT, we also detected enhanced phosphorylation levels by means of Western blotting, suggesting an activation of this kinase. Moreover, analysis of the ROS levels identified an increase in ROS accumulation in renin-b-overexpressing cells. Altogether, our data demonstrate that renin-b overexpression induces the metabolic remodeling of H9c2 cells similar to that seen under oxygen deprivation. This metabolic phenotype exerting so-called aerobic glycolysis is also known as the Warburg effect.
... We previously overexpressed the coding region of renin-b without its 5 untranslated region, which is derived from intron A of the renin gene in H9c2 cells. These cells were protected from necrotic cell death under glucose starvation [9] and from necrotic as well as apoptotic cell death induced by oxygen combined with glucose depletion (OGD) [10]. They also exhibited a switch to a more aerobic glycolysis, known as the Warburg effect, which may be favorable under ischemic conditions [11]. ...
... However, neither the role of the full-length transcript encoding renin-b, nor the role of renin-a, when overexpressed in cardiac cells, are known. To support the hypothesis of an existing endogenously protective renin-b, it was now essential to demonstrate that exactly this transcript, which is found in vivo, exerts the same protective effects as overexpression of exon (2)(3)(4)(5)(6)(7)(8)(9), lacking exon1a and hence its 5 UTR. Therefore, our aims were (1) to investigate the effects of renin-a overexpression in H9c2 cells in general, and (2) to demonstrate that the already observed protective effects of artificial exon (2-9) renin overexpression were still present when the endogenous renin-b was used, including its 5 UTR upstream of exons 2-on necrosis, apoptosis, and the production of reactive oxygen species under starvation conditions such as glucose depletion and anoxia. ...
... An alternative renin transcript, termed renin-b, recently aroused interest as a promising target to protect cardiac cells under ischemia-related conditions. In the rat heart, expression of renin-b but not of renin-a is increased after myocardial infarction [9]. In cardiac H9c2 cells, anoxia and OGD increased the expression of renin-b, but not of renina [10]. ...
Article
Full-text available
A stimulated renin-angiotensin system is known to promote oxidative stress, apoptosis, necrosis and fibrosis. Renin transcripts (renin-b; renin-c) encoding a cytosolic renin isoform have been discovered that may in contrast to the commonly known secretory renin (renin-a) exert protective effects Here, we analyzed the effect of renin-a and renin-b overexpression in H9c2 cardiomyoblasts on apoptosis and necrosis as well as on potential mechanisms involved in cell death processes. To mimic ischemic conditions, cells were exposed to glucose starvation, anoxia or combined oxygen–glucose deprivation (OGD) for 24 h. Under OGD, control cells exhibited markedly increased necrotic and apoptotic cell death accompanied by enhanced ROS accumulation, loss of mitochondrial membrane potential and decreased ATP levels. The effects of OGD on necrosis were exaggerated in renin-a cells, but markedly diminished in renin-b cells. However, with respect to apoptosis, the effects of OGD were almost completely abolished in renin-b cells but interestingly also moderately diminished in renin-a cells. Under glucose depletion we found opposing responses between renin-a and renin-b cells; while the rate of necrosis and apoptosis was aggravated in renin-a cells, it was attenuated in renin-b cells. Based on our results, strategies targeting the regulation of cytosolic renin-b as well as the identification of pathways involved in the protective effects of renin-b may be helpful to improve the treatment of ischemia-relevant diseases.
... Because ren (2)(3)(4)(5)(6)(7)(8)(9) had beneficial effects on survival of cardiac H9c2 cells exposed to glucose depletion 10 , we examined the consequences of anoxia combined with renin downregulation on necrotic and apoptotic cell death. Anoxia alone did not change the rate of necrosis in H9c2 control and scrambled control cells during the time window investigated. ...
... Since it is difficult-if not impossible-to obtain correct data on ANG peptide levels in the cytosol and to differentiate between ANG produced within the cell from that taken up, we took advantage of CH732, a specific blocker of renin activity instead. CH732 is able to enter the cells and to inhibit ANG I generation by renin-a and renin-b as demonstrated previously 8,10,13 . In pIRES control vector transfected cells, CH732 slightly decreased ATP content under normoxic conditions (Fig. 4a) and lactate accumulation under anoxic condition (Fig. 4c). ...
... An alternative renin transcript was discovered in rats and mice 2,3 , as well as in mice overexpressing a human renin transgene 4 . These transcripts encode for a renin isoform, renin-b, that is present in the cytosol and may be cardioprotective instead of harmful under ischemia related conditions 9,10 . The expression of renin-b now provides an important necessity for intra-cytoplasmatic ANG generation (Fig. 5), but also may have angiotensin independent functions (Fig. 5, Model B) (and see below). ...
Article
Full-text available
The renin-angiotensin system is known to regulate blood pressure as well as water- and electrolyte balance. An activated RAS is involved in the development of hypertension and hypertension-related organ damage. Thus, inhibitors of the RAS are protective and markedly increasing the life span of patients. In contrast, renin transcripts have been discovered encoding a cytoplasmatic renin isoform, termed renin-b, which is not harmful but may be even protective. Here we demonstrate that depletion of renin-b encoding transcripts by small interference RNA decreased ATP levels and increased basal necrosis as well as apoptosis rates. Furthermore, renin-b depletion potentiated the anoxia-induced increase of necrosis rates. Vice versa, overexpression of renin-b prevented the anoxia-induced increase of caspase-mediated apoptosis rates. Besides, cells overexpressing renin-b exhibited even reduced mitochondrial mediated apoptosis rates under anoxia, when compared with normoxic conditions, as indicated by Annexin V labeling. However, whereas the protective effect of renin-b on caspase-mediated apoptosis was completely blocked by the renin inhibitor CH732, the effect on mitochondrial-mediated apoptosis was not affected by CH732 at all. From these data we conclude that renin-b overexpression mediates cardioprotective effects under anoxia with respect to mitochondrial induced apoptosis angiotensin-independently, but with respect to caspase induced apoptosis likely in an angiotensin-dependent manner.
... Exon(1A-9)renin transcript levels increase under glucose and/or oxygen depletion in pIRES and ren(2-9) cells. For these experiments, we generated a cell line overexpressing ren (2)(3)(4)(5)(6)(7)(8)(9) mRNA about 10-fold [ren(2-9) cells]. We excluded the so-called "exon1A", since it is non-coding and may have regulatory functions that we thus avoided. ...
... In contrast, exon(1A-9)renin mRNA expression increased in both cell lines after exposure to glucose depletion and anoxia as well as after OGD (Fig. 1A,C). We already demonstrated that glucose depletion increased ren (2)(3)(4)(5)(6)(7)(8)(9) protein in previous studies. We here further confirm that the ren(2-9) protein level was increased in ren (2)(3)(4)(5)(6)(7)(8)(9) transfected cells already prior to the depletion conditions (Fig. 1E). ...
... We already demonstrated that glucose depletion increased ren (2)(3)(4)(5)(6)(7)(8)(9) protein in previous studies. We here further confirm that the ren(2-9) protein level was increased in ren (2)(3)(4)(5)(6)(7)(8)(9) transfected cells already prior to the depletion conditions (Fig. 1E). ...
Article
Full-text available
Although the renin-angiotensin system usually promotes oxidative stress and cell death, renin transcripts have been discovered, whose transcription product may be cardioprotective. These transcripts encode a non-secretory renin isoform that is localized in the cytosol and within mitochondria. Here we tested the hypotheses that cytosolic renin [ren(2-9)] expression promotes cell survival under hypoxia and glucose depletion by preserving the mitochondrial membrane potential (∆Ψm) and mitigating the accumulation of ROS. To simulate ischemic insults, we exposed H9c2 cells to glucose deprivation, anoxia or to combined oxygen-glucose deprivation (OGD) for 24 hours and determined renin expression. Furthermore, H9c2 cells transfected with the empty pIRES vector (pIRES cells) or ren(2-9) cDNA-containing vector [ren(2-9) cells] were analyzed for cell death, ∆Ψm, ATP levels, accumulation of ROS, and cytosolic Ca2+ content. In pIRES cells, expression of ren(1A-9) was stimulated under all three ischemia-related conditions. After OGD, the cells lost their ∆Ψm and exhibited enhanced ROS accumulation, increased cytosolic Ca2+ levels, decreased ATP levels as well as increased cell death. In contrast, ren(2-9) cells were markedly protected from these effects. Ren(2-9) appears to represent a protective response to OGD by reducing ROS generation and preserving mitochondrial functions. Therefore, it is a promising new target for the prevention of ischemia-induced myocardial damage.
... 199 Renin, or angiotensinogenase, is secreted by the granular cells of the kidneys and is found in several isoforms containing 340 amino acids with antagonizing functions. 200 Renin is also considered a hormone because it has a signaling function. The enzyme renin acts on its substrate to form angiotensin II, a universal effector peptide hormone. ...
Article
Full-text available
In the last decade, metagenomic studies have shown the key role of the gut microbiome in maintaining immune and neuroendocrine systems. Malfunction of the gut microbiome can induce inflammatory processes, oxidative stress, and cytokine storm. Dysfunction of the gut microbiome can be caused by short-term (virus infection and other infectious diseases) or long-term (environment, nutrition, and stress) factors. Here, we reviewed the inflammation and oxidative stress in neurodegenerative diseases and coronavirus infection (COVID-19). Here, we reviewed the renin–angiotensin–aldosterone system (RAAS) involved in the processes of formation of oxidative stress and inflammation in viral and neurodegenerative diseases. Moreover, the coronavirus uses ACE2 receptors of the RAAS to penetrate human cells. The coronavirus infection can be the trigger for neurodegenerative diseases by dysfunction of the RAAS. Pharmabiotics, postbiotics, and next-generation probiotics, are considered as a means to prevent oxidative stress, inflammatory processes, neurodegenerative and viral diseases through gut microbiome regulation.
... Studies from Jörg Peters's laboratory have evidenced that cardiac insult triggers renin-b upregulation in the myocardium [37]. Furthermore, myocardial renin-b overexpression results in cardioprotection against ischemic and hypoglycemic conditions [38,39]. Future studies using conditional and tissue-specific models will be required to elucidate the distinct functions of renin-b in different cells including cardiomyocytes. ...
Article
Full-text available
Excessive sodium intake is known to increase the risk for hypertension, heart disease, and stroke. Individuals who are more susceptible to the effects of high salt are at higher risk for cardiovascular diseases even independent of their blood pressure status. Local activation of the renin-angiotensin system (RAS) in the brain, among other mechanisms, has been hypothesized to play a key role in contributing to salt balance. We have previously shown that deletion of the alternative renin isoform termed renin-b disinhibits the classical renin-a encoding preprorenin in the brain resulting in elevated brain RAS activity. Thus, we hypothesized that renin-b deficiency results in higher susceptibility to salt-induced elevation in blood pressure. Telemetry implanted Ren-b Null and wildtype littermate mice were first offered a low salt diet for a week and subsequently a high salt diet for another week. A high salt diet induced a mild blood pressure elevation in both Ren-b Null and wildtype mice, but mice lacking renin-b did not exhibit an exaggerated pressor response. When renin-b deficient mice were exposed to a high salt diet for a longer duration (4 weeks), there was a trend for increased myocardial enlargement in Ren-b Null mice when compared with control mice, but this did not reach statistical significance. Multiple studies have also demonstrated the association of environmental stress with hypertension. Activation of the RAS in the rostral ventrolateral medulla and the hypothalamus is required for stress-induced hypertension. Thus, we next questioned whether the lack of renin-b would result in exacerbated response to an acute restraint-stress. Wildtype and Ren-b Null mice equally exhibited elevated blood pressure in response to restraint-stress, which was similar in mice fed either a low or high salt diet. These studies suggest that mechanisms unrelated to salt and acute stress alter the cardiovascular phenotype in mice lacking renin-b.
... 304 upregulation in the myocardium [34]. Furthermore, myocardial renin-b overexpression 305 results in cardioprotection against ischemic and hypoglycemic conditions [35,36]. Future 306 studies using conditional and tissue-specific models will be required to elucidate the 307 distinct functions of renin-b in different cells including cardiomyocytes. ...
Preprint
Full-text available
Excessive sodium intake is known to increase the risk for hypertension, heart disease, and stroke. Individuals who are more susceptible to the effects of high salt are at higher risk for cardiovascular diseases even independent of their blood pressure status. Local activation of the renin-angiotensin system (RAS) in the brain, among other mechanisms, has been hypothesized to play a key role in contributing to salt balance. We have previously shown that deletion of the alternative renin isoform termed renin-b disinhibits the classical renin-a encoding preprorenin in the brain resulting in elevated brain RAS activity. Thus, we hypothesized that renin-b deficiency results in higher susceptibility to salt-induced elevation in blood pressure. Telemetry implanted Ren-b Null and wildtype littermate mice were first offered a low salt diet for a week and subsequently a high salt diet for another week. A high salt diet induced a mild blood pressure elevation in both Ren-b Null and wildtype mice, but mice lacking renin-b did not exhibit an exaggerated pressor response. When renin-b deficient mice were exposed to a high salt diet for a longer duration (4 weeks), was a trend for increased myocardial enlargement in Ren-b Null mice when compared with control mice. Multiple studies have also demonstrated the association of chronic and acute environmental stress with hypertension. Activation of the RAS in the rostral ventrolateral medulla and the hypothalamus is required for stress-induced hypertension. Thus, we next questioned whether the lack of renin-b would result in exacerbated response to an acute restraint-stress. Wildtype and Ren-b Null mice equally exhibited elevated blood pressure in response to restraint-stress, which was similar in mice fed either a low or high salt diet. These studies highlight a complex mechanism that masks/unmasks roles for renin-b in cardiovascular physiology.
... Overexpression of renin-b in either H9c2 cardiomyoblasts, primary cardiomyocytes, or isolated hearts has been shown to induce cardioprotection against ischemia-induced injury. 14 On the contrary, overexpression of renin-b in rats resulted in increased expression of markers of renal injury in the kidney. 14,15 Thus, whether renin-b is another protective arm of the RAS remains a topic of debate. ...
Article
Full-text available
Several cardiac and renal diseases are attributed to a dysregulation of the renin-angiotensin system. Renin, the rate-limiting enzyme of the renin-angiotensin system, has 2 isoforms. The classical renin isoform (renin-a) encoding preprorenin is mainly confined to the juxtaglomerular cells and released into the circulation upon stimulation. Alternatively, renin-b is predicted to remain intracellular and is expressed in the brain, heart, and adrenal gland. In the brain, ablation of renin-b (Ren-bNull mice) results in increased brain renin-angiotensin system activity. However, the consequences of renin-b ablation in tissues outside the brain remain unknown. Therefore, we hypothesized that renin-b protects from hypertensive cardiac and renal end-organ damage in mice. Ren-bNull mice exhibited normal blood pressure at baseline. Thus, we induced hypertension by using a slow pressor dose of Ang II (angiotensin II). Ang II increased blood pressure in both wild type and Ren-bNull to the same degree. Although the blood pressure between Ren-bNull and wild-type mice was elevated equally, 4-week infusion of Ang II resulted in exacerbated cardiac remodeling in Ren-bNull mice compared with wild type. Ren-bNull mice also exhibited a modest increase in renal glomerular matrix deposition, elevated plasma aldosterone, and a modestly enhanced dipsogenic response to Ang II. Interestingly, ablation of renin-b strongly suppressed plasma renin, but renal cortical renin mRNA was preserved. Altogether, these data indicate that renin-b might play a protective role in the heart, and thus renin-b could be a potential target to treat hypertensive heart disease.
Article
The (pro)renin receptor [(P)RR, Atp6ap2] was initially discovered as a membrane-bound binding partner of prorenin and renin. A soluble (P)RR has additional paracrine effects and is involved in metabolic syndrome and kidney damage. Meanwhile it is clear that most of the effects of the (P)RR are independent of prorenin. In the kidney, (P)RR plays an important role in renal dysfunction by activating proinflammatory and profibrotic molecules. In the brain, (P)RR is expressed in cardiovascular regulatory nuclei and is linked to hypertension. (P)RR is known to be an essential component of the v-ATPase as a key accessory protein and plays an important role in kidney, brain and heart via regulating the pH of the extracellular space and intracellular compartments. V-ATPase and (P)RR together act on WNT and mTOR signalling pathways, which are responsible for cellular homeostasis and autophagy. (P)RR through its role in v-ATPase assembly and function is also important for fast recycling endocytosis by megalin. In the kidney, megalin together with v-ATPase and (P)RR is crucial for endocytic uptake of components in the RAS and their intracellular processing. In the brain, (P)RR, v-ATPases and megalin are important regulators both during development and in the adult. All three proteins are associated with diseases such as XLMR, XMRE, X-linked parkinsonism and epilepsy, cognitive disorders with Parkinsonism, spasticity, intellectual disability, and Alzheimer’s Disease which are characterized by impaired neuronal function and/or neuronal loss. The present review focusses on the relevant effects of Atp6ap2 without assigning them necessarily to the RAS. Mechanistically, many effects can be well explained by the role of Atp6ap2 for v-ATPase assembly and function. Furthermore, application of a soluble (P)RR analogue as new therapeutic option is discussed.
Chapter
The renin-angiotensin system (RAS) constitutes a key hormonal system in the physiological regulation of blood pressure via peripheral and central mechanisms. Dysregulation of the RAS is considered a major factor in the development of cardiovascular pathologies and pharmacologic blockade of this system by the inhibition of angiotensin-converting enzyme (ACE) or antagonism of the angiotensin type 1 receptor (AT1R) is an effective therapeutic regimen. The RAS is now defined as a more complex system composed of different angiotensin peptides with diverse biological actions mediated by distinct receptor subtypes. The classic RAS comprises the ACE-Ang II-AT1R axis that promotes vasoconstriction, water intake, sodium retention and increased oxidative stress, fibrosis, cellular growth, and inflammation. The non-classical or alternative RAS is composed primarily of the ACE2-Ang-(1-7)-AT7R pathway that opposes many actions of the Ang II-AT1R axis. In lieu of the complexity of this system, the chapter discusses the current evidence on the enzymatic cascade of the Ang-(1-7) axis of the RAS regarding the peptidases that contribute to the formation and degradation of the peptide in the circulation and various tissues.
Article
Full-text available
We tested the hypothesis that direct renin inhibition with aliskiren protects against myocardial ischemia/reperfusion (I/R) injury in spontaneously hypertensive rats (SHR), and examined the mechanism by which this occurs. Male SHR were treated (orally, 4 weeks) with saline or aliskiren (30 or 60 mg kg(-1) day(-1)) and subjected to 30 minutes of left anterior descending coronary artery occlusion followed by 6 or 24 hours of reperfusion. Only the higher dose significantly lowered systolic blood pressure, the lower dose causing a smaller apparent lowering that was nonsignificant. Despite this difference in blood pressure-lowering effect, both doses increased the ejection fraction and fractional shortening and reduced myocardial infarct size equally. I/R decreased cardiac expression of phosphatidylinositol 3-kinase (PI3K), phospho-Akt and phospho-endothelial nitric oxide synthase (phospho-eNOS), but increased expression of inducible nitric oxide synthase (iNOS); these changes were all abrogated by aliskiren. Moreover, aliskiren decreased superoxide anion generation and increased cyclic guanosine-3',5'-monophosphate, an index of bioactive nitric oxide, in myocardium. It also decreased the expression of myocardial matrix metalloproteinase-2, matrix metalloproteinase-9, and tissue inhibitor of metalloproteinases-1 (TIMP-1) following I/R. In a Langendorff heart preparation, the detrimental cardiac effects of I/R were abrogated by aliskiren, and these protective effects were abolished by NOS or PI3K inhibition. In a parallel study, although specific iNOS inhibition reduced plasma malondialdehyde and myocardial superoxide anion generation, it did not affect the deleterious effects of I/R on myocardial structure and function. Direct renin inhibition protects against myocardial I/R injury through activation of the PI3K-Akt-eNOS pathway.
Article
Full-text available
The renin-angiotensin (Ang) system regulates multiple physiological functions through Ang II type 1 and type 2 receptors. Prior studies suggest an intracellular pool of Ang II that may be released in an autocrine manner upon stretch to activate surface membrane Ang receptors. Alternatively, an intracellular renin-Ang system has been proposed, with a primary focus on nuclear Ang receptors. A mitochondrial Ang system has not been previously described. Here we report that functional Ang II type 2 receptors are present on mitochondrial inner membranes and are colocalized with endogenous Ang. We demonstrate that activation of the mitochondrial Ang system is coupled to mitochondrial nitric oxide production and can modulate respiration. In addition, we present evidence of age-related changes in mitochondrial Ang receptor expression, i.e., increased mitochondrial Ang II type 1 receptor and decreased type 2 receptor density that is reversed by chronic treatment with the Ang II type 1 receptor blocker losartan. The presence of a functional Ang system in human mitochondria provides a foundation for understanding the interaction between mitochondria and chronic disease states and reveals potential therapeutic targets for optimizing mitochondrial function and decreasing chronic disease burden with aging.
Article
Full-text available
Angiotensin-II (Ang-II) from extracardiac sources and intracardiac synthesis regulates cardiac homeostasis, with mitogenic and growth-promoting effects largely due to altered gene expression. Here, we assessed the possibility that angiotensin-1 (AT1R) or angiotensin-2 (AT2R) receptors on the nuclear envelope mediate effects on cardiomyocyte gene expression. Immunoblots of nucleus-enriched fractions from isolated cardiomyocytes indicated the presence of AT1R and AT2R proteins that copurified with the nuclear membrane marker nucleoporin-62 and histone-3, but not markers of plasma (calpactin-I), Golgi (GRP-78), or endoplasmic reticulum (GM130) membranes. Confocal microscopy revealed AT1R and AT2R proteins on nuclear membranes. Microinjected Ang-II preferentially bound to nuclear sites of isolated cardiomyocytes. AT1R and AT2R ligands enhanced de novo RNA synthesis in isolated cardiomyocyte nuclei incubated with [alpha-(32)P]UTP (e.g. 36.0 +/- 6.0 cpm/ng of DNA control versus 246.4 +/- 15.4 cpm/ng of DNA Ang-II, 390.1 +/- 15.5 cpm/ng of DNA L-162313 (AT1), 180.9 +/- 7.2 cpm/ng of DNA CGP42112A (AT2), p < 0.001). Ang-II application to cardiomyocyte nuclei enhanced NFkappaB mRNA expression, a response that was suppressed by co-administration of AT1R (valsartan) and/or AT2R (PD123177) blockers. Dose-response experiments with Ang-II applied to purified cardiomyocyte nuclei versus intact cardiomyocytes showed greater increases in NFkappaB mRNA levels at saturating concentrations with approximately 2-fold greater affinity upon nuclear application, suggesting preferential nuclear signaling. AT1R, but not AT2R, stimulation increased [Ca(2+)] in isolated cardiomyocyte nuclei. Inositol 1,4,5-trisphosphate receptor blockade by 2-aminoethoxydiphenyl borate prevented AT1R-mediated Ca(2+) release and attenuated AT1R-mediated transcription initiation responses. We conclude that cardiomyocyte nuclear membranes possess angiotensin receptors that couple to nuclear signaling pathways and regulate transcription. Signaling within the nuclear envelope (e.g. from intracellularly synthesized Ang-II) may play a role in Ang-II-mediated changes in cardiac gene expression, with potentially important mechanistic and therapeutic implications.
Article
An alternative transcript of the rat renin gene was recently characterized in the adrenal gland, in addition to the known messenger RNA (mRNA) coding for preprorenin. In the alternative transcript, exon 1 is replaced by exon 1A, a domain originating in intron 1. The reading frame of this mRNA, termed exon 1A-renin transcript, codes for a truncated prorenin that presumably remains intracellular, in contrast to preprorenin, which is targeted to the secretory pathway by its prefragment. We here demonstrate the tissue-specific regulation of expression of both transcripts by RT and PCR. In many tissues both transcripts are present, for example in the adrenal gland, spleen, liver, and hypothalamus. In some organs, however, only one of the renin mRNAs is found. In the kidney only the full-length mRNA coding for preprorenin is detected. In the heart exclusively the exon 1A-mRNA is expressed, but not the preprorenin transcript. After myocardial infarction, which is known to activate the intracardiac renin-angiotensin system, expression of exon 1A-renin mRNA in the left ventricle was stimulated about 4-fold, compared with that in sham-operated animals, whereas no mRNA corresponding to preprorenin was detectable. These findings may have implications for the current concepts of local extrarenal renin-angiotensin systems, as they provide the molecular basis for a possible intracellular function of renin and exclude a role for locally produced secretory renin in the heart.
Article
Angiotensin-converting enzyme inhibitors and angiotensin AT1 receptor blockers reduce myocardial ischemia-reperfusion injury via bradykinin B2 receptor- and angiotensin AT2 receptor-mediated mechanisms. The renin inhibitor aliskiren increases cardiac tissue kallikrein and bradykinin levels. In the present study, we investigated the effect of aliskiren on myocardial ischemia-reperfusion injury and the roles of B2 and AT2 receptors in this effect. Female Sprague-Dawley rats were treated with aliskiren (10 mg/kg per day) and valsartan (30 mg/kg per day), alone or in combination, together with the B2 receptor antagonist icatibant (0.5 mg/kg per day) or the AT2 receptor antagonist PD123319 (30 mg/kg per day), for 4 weeks before myocardial ischemia-reperfusion injury. Aliskiren increased cardiac bradykinin levels and attenuated valsartan-induced increases in plasma angiotensin II levels. In vehicle-treated rats, myocardial infarct size (% area at risk, mean±SEM, n=7-13) was 43±3%. This was reduced to a similar extent by aliskiren, valsartan, and their combination to 24±3%, 25±3%, and 22±2%, respectively. Icatibant reversed the cardioprotective effects of aliskiren and the combination of aliskiren plus valsartan, but not valsartan alone, indicating that valsartan-induced cardioprotection was not mediated by the B2 receptor. PD123319 reversed the cardioprotective effects of aliskiren, valsartan, and the combination of aliskiren plus valsartan. Aliskiren protects the heart from myocardial ischemia-reperfusion injury via a B2 receptor- and AT2 receptor-mediated mechanism, whereas cardioprotection by valsartan is mediated via the AT2 receptor. In addition, aliskiren attenuates valsartan-induced increases in angiotensin II levels, thus preventing AT2 receptor-mediated cardioprotection by valsartan.
Article
Renin is known as a secretory glycoprotein produced, stored and released by the kidney. Renin cleaves angiotensin (ANG) I from angiotensinogen, which is further cleaved to ANG II by ANG-converting enzyme. ANG II acts via specific receptors located at the cell membrane. Recently renin transcripts have been discovered which encode a cytosolic protein that cannot be secreted. These transcripts are derived from the same renin gene but probably use another promoter located within intron A. Whereas the kidney exclusively expresses the transcript encoding secretory renin, other tissues additionally or exclusively express transcripts encoding cytosolic renin. The cytosolic renin protein exerts functions different and even opposite to those of secretory renin. Whereas secretory renin increases necrotic death rates of cardiac cells, the cytosolic renin isoform even protects cells from necrotic death. This review describes the properties of cytosolic renin and its cellular functions and discusses possible mechanisms of actions particularly in the light of the discovery of direct nuclear and mitochondrial effects of ANG' s.
Article
Ischemic heart disease (IHD) is the leading cause of death and disability worldwide. The major pathological consequences of IHD arise from the detrimental effects of acute ischemia-reperfusion injury (IRI) on the myocardium. Therefore, in order to improve clinical outcomes in patients with IHD, novel therapeutic strategies are required to protect the myocardium from acute IRI and preserve cardiac function (cardioprotection). In this regard, endogenous cardioprotective strategies such as ischemic preconditioning (IPC), ischemic postconditioning (IPost) and remote ischemic conditioning (RIC) may provide novel approaches for protecting the heart in clinical settings in which the patient experiences acute myocardial IRI. In this review article, we provide an overview of these endogenous cardioprotective strategies with respect to the pre-clinical experimental literature, exploring their major characteristics and underlying signaling mechanisms. The application of these therapeutic strategies in the clinical setting for potential patient benefit is reviewed in another article in this special issue.
Article
Myocardial infarct size is a major determinant of prognosis. Ischaemic preconditioning with brief coronary occlusion and reperfusion before a sustained period of coronary occlusion with reperfusion delays infarct development. Ischaemic postconditioning uses repetitive brief coronary occlusion during early reperfusion of myocardial infarction and reduces infarct size. Remote ischaemic preconditioning uses brief ischaemia and reperfusion of a distant organ to protect the myocardium. These conditioning protocols recruit a complex signal cascade of sarcolemmal receptor activation, intracellular enzyme activation, and ultimately mitochondrial stabilisation and inhibition of death signalling. Conditioning protocols have been successfully used in patients undergoing elective coronary revascularisation and reperfusion after acute myocardial infarction. Pharmacological recruitment of cardioprotective signalling has also been used to reduce infarct size, but so far without prognostic benefit. Outcomes of cardioprotection are affected by age, sex, comorbidities, and drugs, but also by technical issues related to determination of infarct size and revascularisation procedure.