ArticlePDF Available

Dipeptidyl peptidase 3 modulates the renin–Angiotensin system in mice

Authors:

Abstract and Figures

Dipeptidyl peptidase 3 (DPP3) is a zinc-dependent hydrolase involved in degrading oligopeptides with 4–12 amino acid residues. It has been associated with several pathophysiological processes, including blood pressure regulation, pain signaling, and cancer cell defense against oxidative stress. However, the physiological substrates and the cellular pathways that are potentially targeted by DPP3 to mediate these effects remain unknown. Here, we show that global DPP3 deficiency in mice (DPP3-/-) affects the renin–angiotensin system (RAS). LC-MS–based profiling of circulating angiotensin peptides revealed elevated levels of angiotensin II, III, IV, and 1–5 in DPP3-/- mice, while blood pressure, renin activity, and aldosterone levels remained unchanged. Activity assays using the purified enzyme confirmed that angiotensin peptides are substrates for DPP3. Aberrant angiotensin signaling was associated with substantially higher water intake and increased renal reactive oxygen species (ROS) formation in the kidneys of DPP3-/- mice. The metabolic changes and altered angiotensin levels observed in male DPP3-/- mice were either absent or attenuated in female DPP3-/- mice, indicating sex-specific differences. Taken together, our observations suggest that DPP3 regulates the RAS pathway and water homeostasis by degrading circulating angiotensin peptides.
Content may be subject to copyright.
1
Dipeptidyl peptidase 3 modulates the reninangiotensin system in mice
Shalinee Jha, Ulrike Taschler, Oliver Domenig3, Marko Poglitsch3, Benjamin Bourgeois4, Marion
Pollheimer5, Lisa M. Pusch2, Grazia Malovan1, Saša Frank4, Tobias Madl4,6, Karl Gruber2, Robert
Zimmermann2,6 and Peter Macheroux1*
1Institute of Biochemistry, Graz University of Technology, NAWI Graz, Petersgasse 12, 8010 Graz,
Austria
2Institute of Molecular Biosciences, University of Graz, NAWI Graz, Heinrichstrasse 31 and
Humboldtstrasse 50, 8010 Graz, Austria
3Attoquant Diagnostics GmbH, Campus Vienna Biocenter 5, 1030 Vienna, Austria
4Gottfried Schatz Research Center, Division of Molecular Biology and Biochemistry,
Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria
5Diagnostic & Research Institute of Pathology, Medical University of Graz, Neue Stiftingtalstrasse 6,
8010 Graz, Austria
6BioTechMed Graz, 8010 Graz, Austria
*to whom correspondence should be addressed:
Prof. Dr. Peter Macheroux, Graz University of Technology, Institute of Biochemistry, Petersgasse
12/2, A-8010 Graz, Austria; Tel.: +43-316-873 6450; Fax: +43-316-873 6952;
Email: peter.macheroux@tugraz.at
§These authors have contributed equally to this work.
Running title: Physiological role of DPP3
Keywords: angiotensin II, dipeptidyl peptidase 3 (DPP3), metalloprotease, mouse, oxidative stress,
peptidase, renal physiology, renin angiotensin system, kidney function, sex-specific difference
https://www.jbc.org/cgi/doi/10.1074/jbc.RA120.014183The latest version is at
JBC Papers in Press. Published on June 16, 2020 as Manuscript RA120.014183
at FACHBIBLIOTHEK CHEMIE on June 16, 2020http://www.jbc.org/Downloaded from
2
Abstract
Dipeptidyl peptidase 3 (DPP3) is a zinc-
dependent hydrolase involved in degrading
oligopeptides with 412 amino acid residues. It
has been associated with several
pathophysiological processes, including blood
pressure regulation, pain signaling, and cancer
cell defense against oxidative stress. However,
the physiological substrates and the cellular
pathways that are potentially targeted by DPP3
to mediate these effects remain unknown.
Here, we show that global DPP3 deficiency in
mice (DPP3-/-) affects the reninangiotensin
system (RAS). LC-MSbased profiling of
circulating angiotensin peptides revealed
elevated levels of angiotensin II, III, IV, and
15 in DPP3-/- mice, while blood pressure,
renin activity, and aldosterone levels remained
unchanged. Activity assays using the purified
enzyme confirmed that angiotensin peptides
are substrates for DPP3. Aberrant angiotensin
signaling was associated with substantially
higher water intake and increased renal
reactive oxygen species (ROS) formation in
the kidneys of DPP3-/- mice. The metabolic
changes and altered angiotensin levels
observed in male DPP3-/- mice were either
absent or attenuated in female DPP3-/- mice,
indicating sex-specific differences. Taken
together, our observations suggest that DPP3
regulates the RAS pathway and water
homeostasis by degrading circulating
angiotensin peptides.
Introduction
Dipeptidyl peptidase 3 (DPP3, EC 3.4.14.4) is
a metalloprotease that specifically cleaves
dipeptides at the N-terminus of peptides with 4
to 12 amino acids. It is ubiquitously expressed
in both, prokaryotes and eukaryotes. DPP3 is
part of the central human proteome, i.e. it
belongs to a set of proteins ubiquitously and
abundantly expressed in all human cells (1).
The crystal structures of bacterial, yeast, and
human DPP3 have been reported (24). All
these structures are composed of an upper and
a lower domain separated by a wide cleft
which has been shown to be the substrate
binding site (4, 5). The conserved (HEXXGH)
and (EECRAE/D) motifs are part of the upper
domain, and are involved in the coordination
of a catalytically essential zinc ion in the
binding site (3).
A variety of small bioactive peptides, such as
met-enkephalin and angiotensin (I and II), are
substrates of DPP3 although the full range of
substrate peptides remains undefined (1, 5).
Consequently, DPP3 has been implicated in
pain modulation (6, 7) and blood pressure
regulation (8, 9). In addition, DPP3 exhibits a
moonlighting activity in the kelch-like ECH-
associated protein 1 (Keap1)-nuclear factor
erythroid 2-related factor 2 (Nrf2) signaling
pathway, which appears to play a role in stress
responses through transcriptional regulation of
the antioxidant response element (ARE) (10).
Despite the structural and biochemical
evidence indicating an intriguing involvement
of DPP3 in peptide processing and signaling as
well as in the response to oxygen stress, its
physiological role and potential involvement in
disease-related processes is currently
unknown.
Recently, it was reported that adult DPP3
knockout mice exhibited a growth defect,
increased bone loss and significantly elevated
bone marrow cellularity. Deletion of DPP3
also resulted in oxidative stress and alterations
of bone microenvironment favoring osteoclast
over osteoblast lineage. The osteoclasts
showed increased reactive oxygen species
(ROS) production, which made them prone to
apoptosis (11). A previous study further
established that DPP3 administration to
Angiotensin II (Ang II)-induced hypertensive
mice could significantly diminish systolic
blood pressure, cardiac hypertrophy, and
myocardial fibrosis in an extent at par with the
effect of the angiotensin receptor blocker
candesartan. It was also observed that DPP3
effectively reduced urine albumin excretion,
kidney damage, and the renal protein levels of
the pro inflammatory molecule monocyte
chemo-attractant protein-1 and the pro-
coagulant platelet activator inhibitor (9). Taken
together, the enzyme’s ability to degrade
various bioactive peptides, DPP3 may have
complex effects and influence basic
physiological processes, particularly those
affecting cellular metabolism and oxidative
stress.
Ang II, which is the most prominent substrate
reported for DPP3, is the principal effector of
at FACHBIBLIOTHEK CHEMIE on June 16, 2020http://www.jbc.org/Downloaded from
3
the renin-angiotensin system (RAS). RAS
plays a pivotal role in the pathophysiological
modulation of renal and cardiovascular
processes (12, 13). Ang II regulates
vasoconstriction and is responsible for
maintaining homeostasis in the heart and
kidney (14). In addition, Ang II is a potent
stimulator of NAD(P)H oxidase, which
augments formation of ROS in various tissues.
Ang II-mediated ROS production has been
associated with cell growth, apoptosis, cell
migration, and expression of inflammatory and
extracellular matrix genes (15). An imbalance
between the production of ROS and the
antioxidant defense to eliminate these toxic
intermediates leads to oxidative stress. There is
a plethora of evidence demonstrating the
importance of oxidative stress in Ang II-
induced metabolic disorders like hypertension,
diabetes mellitus, and chronic kidney disease
(1619). Although blockade of the RAS is the
most commonly adopted strategy to slow
progression of cardiovascular and associated
renal diseases, a better understanding of the
novel aspects of the RAS is of paramount
importance for the development of innovative
therapies that target pathologies inflicted by
anomalies of this pathway.
In the present study, we attempted to elucidate
the physiological role of DPP3 in the RAS
system through characterization of DPP3
knockout mice (DPP3-/-). Our observations
suggest that DPP3 regulates the RAS pathway
and water homeostasis by degrading
circulating angiotensin peptides. Interestingly,
the lack of DPP3 affected only the phenotype
of male mice, with the effects either being
absent or much weaker in female mice. This
sex-specific difference points at a link between
the endocrine system and the physiological
role of DPP3. The characterization of DPP3 in
this study establishes that it has strong
metabolic implications through the modulation
of the RAS pathway, a property that could be
useful in the management of several
cardiovascular and related metabolic
pathologies.
Results
Generation and gross characterization of
DPP3 knockout mice
To investigate the function of DPP3 in vivo,
we generated mice globally lacking DPP3.
Mice were generated using ES cells from
EUCOMM containing a β-galactosidase
cassette (lacZ) and a promotor-driven selection
cassette (neo) between exon 5-6 of the DPP3
gene. The selection cassettes as well as exon 6
of DPP3 were flanked by loxP sites. Mice
bearing the targeted allele were crossed with
transgenic mice expressing Cre-recombinase
under the control of a cytomegalovirus (CMV)
promotor resulting in deletion of neo and exon
6 (Figure 1A). DPP3 was detected by Western
blotting in most investigated tissues of wild-
type controls (DPP3+/+), but not in tissue
lysates of DPP3-/- mice (Figure 1B). A
comparison of DPP3 activity in various tissues
of DPP3+/+ and DPP3-/- mice using the artificial
substrate Arg-Arg-2-naphthylamide clearly
demonstrated blunted activity in DPP3-/- tissue
lysates (Figure 1C).
In accordance with a previously published
study (11), gross characterization revealed that
male DPP3-/- mice exhibit lower body weight
(Figure 2A) and less fat mass than litter-mates
(Figure 2B). Food and water consumption as
well as energy expenditure and spontaneous
locomotor activity were monitored in
metabolic cages over a period of 150 h.
Cumulative analysis revealed a slight increase
in food intake in male DPP3-/- mice (Figure
2C) due to increased food consumption during
the dark phase (Figure 2F). Cumulative water
intake was significantly elevated (Figure 2D)
due to a 27% and 46% increase in drinking
during the light and dark phase respectively
(Figure 2G). Cumulative (Figure 2E) and daily
(Figure 2H) locomotor activity remained
unchanged. As shown in Figure 3A-F, oxygen
consumption and carbon-dioxide production
were comparable between genotypes, while the
respiratory exchange ratio (RER) was slightly
increased in the knockout mice during the dark
phase. We also calculated energy expenditure
(EE) based on the amount of oxygen consumed
and carbon dioxide produced, using the
formula: EE (kJ per day) = 15.818*VO2 +
5.176*VCO2/1000*24 (20) and was found to
remain unaltered between the genotypes
(DPP3+/+, 42.6 ± 2.3 kJ per day vs. DPP3-/-,
43.7 ± 2.8 kJ per day; total energy
expenditure).
at FACHBIBLIOTHEK CHEMIE on June 16, 2020http://www.jbc.org/Downloaded from
4
Mice consume substantially lower amounts of
water after food deprivation. To investigate if
drinking behavior of male DPP3-/- mice was
also altered upon food restriction, we
monitored water consumption during a 13h
fasting period. Under these conditions, water
intake of DPP3-/- mice was ~ 4-fold higher as
compared to DPP3+/+ mice suggesting that
DPP3-deficiency is associated with polydipsia
in male animals (Figure 4).
DPP3 acts as a modulator of the RAS
Our observations indicate that DPP3 is
involved in the regulation of water homeostasis
and recently published data suggested a role of
DPP3 in angiotensin degradation (9, 21).
Accordingly, we generated a serum “RAS-
Fingerprint” consisting of 10 different
angiotensin peptides using ultra-pressure-
liquid chromatography-tandem mass
spectrometry (LC-MS/MS). Serum analysis
revealed that male DPP3-/- mice had higher
concentrations of most angiotensin
metabolites. As shown in Figure 5A, the
concentration of Ang II was twice as high as in
wild-type mice. Interestingly, downstream
peptides, in particular Ang (1-5), Ang III, and
Ang IV accumulated 5.4-, 4.2-, and 5.3-fold,
respectively, suggesting that DPP3 has
multiple natural substrates among angiotensin
metabolites and that DPP3 deletion leads to
perturbation of the entire RAS in males. Serum
aldosterone levels (Figure 5B) and serum renin
activity (Figure 5C) remained unchanged.
Deletion of DPP3 enhances oxidative stress in
male mice
Ang II is known to promote ROS production in
kidney, and therefore we determined the
generation of reactive oxygen intermediates
using 2′,7′-dichlorodihydrofluorescein
diacetate (H2DCFDA). DPP3 deletion led to a
significantly enhanced fluorescence signal
from the ROS reporter dye H2DCFDA in the
kidney lysates prepared from male mice
(Figure 6A). Accumulation of ROS in the
DPP3-/- kidneys also triggered a trend towards
higher malondialdehyde (MDA) levels, a
marker of lipid peroxidation (Figure 6B).
Catalase activity was significantly increased in
kidney homogenates of male DPP3-/- mice,
indicating increased generation of H2O2
(Figure 6C). We also observed that despite this
increase in renal ROS, the kidney morphology
was not significantly altered between the
genotypes (Figure 7). We observed normal
glomeruli with mesanglial normocellularity
and thin basement of the capillary loops.
Moreover, renal tubules and interstitium
appeared normal, lacking signs of
inflammation or fibrosis.
DPP3 deficiency does not affect blood
pressure
The extensive changes in the RAS
accompanied by increased oxidative stress
levels in kidneys of DPP3-/- mice prompted us
to assess potential effects on blood pressure,
which is one of the major physiological output
parameters of the RAS. Toward that end, we
measured blood pressure in 18-22 weeks old
DPP3+/+ and DPP3-/- mice by the tail-cuff
method. There was no significant difference
between DPP3+/+ and DPP3-/- mice, both for the
systolic (DPP3+/+, 145.6 ± 12.1 mm Hg vs.
DPP3-/-, 134.2 ± 22.7 mm Hg) and diastolic
(DPP3+/+, 112.6 ± 12.4 mm Hg vs. DPP3-/-,
99.0 ± 22.5 mm Hg) blood pressure (Figure 8).
Purified DPP3 catalyzes the turnover of
multiple angiotensin peptides
The pleiotropic effect of DPP3 deletion on
angiotensin peptides suggests that the enzyme
does not only accept Ang II as a substrate but
may also degrade other peptides in this
pathway. Thus, we investigated the activity of
DPP3 against different angiotensin peptides.
Kinetic parameters of angiotensin peptides
were obtained by single injection calorimetry
measurements that are based on the exothermal
hydrolysis of peptide bonds by purified
recombinant human DPP3 which shares 93%
sequence identity with the mouse DPP3.
Figure 9 displays the curves for the rate of the
reaction, calculated from integrated raw data
using the Enzyme Kinetics - Single Injection
fitting model. The graphs show the rate of
angiotensin conversion as a function of its
concentration. The insets depict the raw data of
heat change due to the conversion of substrate
peptides. The dependence of the reaction rate
on angiotensin concentration followed typical
MichaelisMenten kinetics, and the parameters
- enthalpy heat change (ΔH), turnover number
(kcat) and Michaelis-Menten constant (KM) -
were found by the fitting model in the
MicroCal PEAQ-ITC Analysis Software
(Table 1). Among the six angiotensin peptides
at FACHBIBLIOTHEK CHEMIE on June 16, 2020http://www.jbc.org/Downloaded from
5
tested, Ang I, Ang II, Ang (1-5), and Ang (1-7)
showed an exothermic reaction, indicating
their turnover by purified hDPP3 (Figure 9A-
C). However, in the case of Ang I, data
extraction was not possible due to the weak
exothermic signal observed, indicating that
Ang I is a poor substrate of DPP3. In contrast,
Ang III and Ang IV displayed both
endothermic and exothermic behavior, a
characteristic observed for slow substrates like
the peptide tynorphin and its derivatives
(Figure 9D-F) (5). These experiments clearly
confirm that DPP3 does not only act on Ang II
but also efficiently hydrolyzes Ang (1-5) and
Ang (1-7).
DPP3-deficiency has only minor impact on
the phenotype of female mice
In contrast to male mice, female DPP3-/- mice
did not show any significant difference in body
weight (Figure 10A) or body mass composition
(Figure 10B). Moreover, food intake (Figure
10C) and water consumption (Figure 10D)
were not different between the genotypes.
Most importantly, serum levels of Ang II
remained unaltered in female DPP3-/- mice as
compared to wild-type, while the levels of Ang
III, Ang IV, and Ang (1-5) were 2.4-, 2-, and
7.5-fold higher, respectively (Figure 10E).
Contrary to male DPP3-/- mice, the amount of
ROS generated was significantly lower in the
kidney lysates of DPP3-/- female mice,
indicating sex-specific differences (Figure
10F).
Discussion
In this study, we demonstrate that deletion of
DPP3 in mice caused widespread
physiological and biochemical changes. In
addition to the previously observed alterations
in body weight and bone morphology (11), the
lack of DPP3 clearly affects the RAS pathway,
which is associated with increased water
consumption. This is characteristic for mouse
models lacking genes associated with the RAS
like ACE or angiotensinogen, as they have
diminished ability to concentrate urine due to
impaired renal development (22, 23).
However, we found no obvious changes in
kidney morphology, indicating that, at least in
the young mice investigated in this study, renal
function is normal.
Male DPP3-/- mice displayed increased levels
of equilibrium angiotensin peptides including
Ang II as well as a specific qualitative shift in
angiotensin metabolite profiles, characterized
by selective and profound increases of Ang 1-
5, Ang III, and Ang IV. These profound
differences in angiotensin profiles indicate that
a general up-regulation of the RAS at the level
of renin can be excluded as an underlying
cause for the observed Ang II increase. Ang II
is an integral part of the RAS and mediates
various physiological responses. High Ang II
levels increases reactive oxygen species and
oxidative stress, and depresses mitochondrial
energy metabolism (15, 16, 18, 19, 24). There
are several reports suggesting that high
circulating Ang II concentration is a stimulus
for thirst in a variety of species, which is
consistent with our findings in male DPP3-/-
mice (14, 25, 26). Since Ang II (16, 18, 24, 27,
28) mediates ROS production, which can
contribute to oxidative stress, we measured
putative stress markers in the kidneys of mice.
Congruously, the level of ROS generation and
catalase activity were elevated in the
knockouts. These results confirm our
hypothesis that increased Ang II creates
oxidative stress in the male DPP3-/- mice.
In addition to regulating stress response via
modulation of Ang II, it is likely that DPP3 is
part of endogenous defense system against
oxidative stress. DPP3 is known to promote
nuclear migration of transcription factor Nrf2
by displacing Keap1 (10, 29). The Keap1-Nrf2
pathway is a key regulator of cellular stress
response caused by ROS. Under basal
conditions, Nrf2 is bound to Keap1. Upon
activation by oxidative stressors, Nrf2
translocates to the nucleus of the cell where it
activates the ARE, thereby regulating the
transcription of genes responsible for
antioxidant and anti-inflammatory defense
(3033). It has been reported that
overexpression of DPP3 could potently
activate the ARE in neuroblastoma cells (IMR-
32 cells) leading to increased expression levels
of NAD(P)H: quinone oxidoreductase 1
(NQO1), a phase II detoxifying enzyme
regulated by ARE. It was also found that DPP3
overexpression efficiently attenuated the toxic
effects of H2O2 and rotenone, demonstrating
the cytoprotective effect of DPP3 against
oxidative stress (34). Furthermore, it was
demonstrated that the DPP3 plasma levels are
at FACHBIBLIOTHEK CHEMIE on June 16, 2020http://www.jbc.org/Downloaded from
6
associated with the survival rate of patients
suffering from sepsis (35), cardiogenic shock
(36), heart failure (37), and acute kidney injury
(38). Our results are also consistent with a
recent study, which reported that lack of DPP3
leads to impaired bone homeostasis and
enhanced oxidative stress in the bone tissue
(11), indicating an ubiquitous antioxidant
activity of DPP3.
Increase in the Ang II levels leads to
vasoconstriction and thereby, severe
hypertension (17, 19, 27). However, both male
and female DPP3-/- mice showed no change in
the blood pressure using tail-cuff method under
normal dietary conditions. As measuring blood
pressure using the tail-cuff method has certain
limitations (e.g. stress response of mice),
further studies will be necessary to investigate
a potential fine tuning of blood pressure
regulation in DPP3-/- mice. The unchanged
blood pressure also points towards the
involvement of the cardioprotective arm of
RAS as a compensatory mechanism (8). In
vitro studies using recombinant human DPP3
additionally identified Ang (1-7) and Ang (1-5)
as substrates of the enzyme. Although the level
of Ang (1-7) was not significantly increased in
the knockout, it is interesting to note that it is
the main metabolite of the alternate RAS and
plays an important role as physiological
antagonist of Ang II, having vasodilatory and
antihypertensive properties (8). Ang (1-7) is
produced by carboxy- or endopeptidases like
neutral endopeptidase (NEP), prolyl
endopeptidase (PEP), angiotensin converting
enzyme 2 (ACE 2), or prolyl carboxypeptidase
(PCP) from Ang I and Ang II and is
metabolized to Ang (2-7) and Ang (3-7) by
aminopeptidases and to Ang (1-5) by ACE. In
contrast to Ang (1-7), Ang (1-5) showed a very
pronounced change between wild-type and
knockout mice exhibiting a more than five-fold
higher concentration in the latter. The specific
increase of Ang (1-5) in DPP3-/- mice points to
a clear preference of DPP3 towards Ang (1-5)
over Ang (1-7). It was shown that Ang (1-5)
stimulated the secretion of atrial natriuretic
peptide (ANP) via Mas receptor, a mode of
action similar to that of Ang (1-7) (39). The
ANP system is a hormonal system that
participates in the regulation of body fluid and
electrolyte balance. It acts antagonistically to
the RAS and causes anti-hypertension and anti-
oxidative stress (40, 41). It is conceivable that
Ang (1-5) also plays a role as a vasodilatory
and antihypertensive peptide.
Female DPP3-/- mice displayed a less
pronounced phenotype in comparison to male
mice, indicating sex-specific differences.
Accumulating evidence suggests that there are
sex differences in the tissue expression and
activity of several RAS components, with the
female sex hormone, estrogen, downregulating
Ang II and upregulating Ang (1-7) pathways
(42). It was shown that in C57BL/6J mice, the
Ang II‐induced increase in blood pressure is
greater in males than in females (42). Also,
healthy men had greater pressor and renal
vasoconstrictor responses to acute Ang II
infusion compared to women (43). Similarly,
chronic Ang II infusion induced hypertension
in male but not female mice (44, 45), possibly
due to a shift in the balance from Ang II
towards Ang (1-7) pathways because of
estrogen-mediated protection (46). In high-fat
fed C57BL/6J mice, females maintained
circulating Ang (1-7) levels and were protected
from hypertension and metabolic
complications induced by Ang II (47). These
studies are in line with our observations from
the serum RAS-fingerprint which show that the
female DPP3+/+ and DPP3-/- mice had 6-, and
9-fold higher Ang (1-7) respectively, than the
male mice. For Ang (1-5), the levels were 2-,
and 3-fold higher than in male DPP3+/+ and
DPP3-/- mice, respectively. This is indicative of
an upregulation of the protective RAS arm in
female mice.
On a similar note, numerous studies have
discussed the association of DPP3 to estrogen.
It was reported that 17ß-estradiol (E2), the
predominant estrogen present in the serum,
influences the expression level of DPP3 in
vivo. The hepatic DPP3 levels were found to
be markedly reduced following ovariectomy in
16-weeks old CBA/H mice and E2
administration abolished this effect and
increased DPP3 protein expression (48). It was
also found that DPP3 is instrumental in
estrogen-mediated protection against oxidative
stress in female CBA/H mice (49). DPP3
accumulated in the nucleus in liver tissue
lysates of healthy female mice exposed to
hyperoxia, at a level comparable to the nuclear
accumulation of Nrf2. Further, the combined
induction of hyperoxia and E2 administration
had a synergistic effect on the nuclear
at FACHBIBLIOTHEK CHEMIE on June 16, 2020http://www.jbc.org/Downloaded from
7
accumulation of DPP3. In ovariectomized
females exposed to hyperoxia,
supplementation of E2 enhanced DPP3 levels,
accompanied by an upregulation of other
cytoprotective proteins like sirtuin-1 and heme
oxygenase-1, resulting in attenuated oxidative
stress (49). Lack of DPP3 was found to
augment bone loss caused by estrogen
deprivation in the ovariectomized mouse
model of human postmenopausal osteoporosis
(11). This interplay between DPP3, RAS, and
estrogen demands additional studies, however,
it is very likely that the female DPP3-/- mice are
protected from oxidative stress due to the
presence of estrogen. The male DPP3-/- mice on
the other hand show an aggravated response to
Ang II-induced oxidative stress due to this lack
of antioxidant and cytoprotective function.
In summary, the generation and
characterization of a mouse model with global
deletion of DPP3 has revealed a significant
perturbation of the levels of peptides in the
RAS. The changes in peptide levels were
found to be associated with polydipsia and
augmented levels of ROS. Our findings
identify DPP3 as a pleiotropic and sex-specific
modulator of RAS and emphasize its role in
oxidative stress response.
Experimental procedures
Ethics Statement
All animal experiments were approved by the
Austrian Federal Ministry for Science,
Research, and Economy (protocol number
BMWF-66.007/7-ll/3b/), the ethics committee
of the University of Graz, and conducted in
compliance with the council of Europe
Convention (ETS 123).
Animals and generation of DPP3 knockout
mice
All studies were conducted in age-matched
DPP3-/- and wild-type control male and female
mice on C56BL/6J background. Unless stated
otherwise, the results describe the effects of
DPP3 deletion on male mice. Mice were bred
and maintained at regular housing
temperatures (23 ± 1 °C) and 14-h light/10-h
dark cycle. Animals had ad libitum access to
water and chow diet (4.5% fat, 34% starch,
5.0% sugar, and 22.0% protein; Ssniff
Spezialdiaeten). Breeding and genotyping were
done according to standard procedures. For
generation of DPP3-/- mice, targeted mutant ES
cells were obtained from EUCOMM and
injected into blastocysts of C57BL/6J mice.
Chimeric animals with a high degree of coat
color chimerism were bred with C57BL/6J
mice. The construct containing a β-
galactosidase cassette (lacZ) and a promotor-
driven selection cassette (neo) was inserted
into the DPP3 gene. Additionally, the
construct contained two flippase recognition
target (FRT) sites for flippase recombination
enzyme (FLP)-mediated recombination
flanking lacZ and neo. The selection cassette
and exon 6 (essential for DPP3 function),
flanked by loxP sites, were removed by
breeding with transgenic C57BL/6J mice
expressing cre-recombinase under the control
of a cytomegalovirus (CMV) promotor (CMV-
Cre). Cre-lox recombination resulted in
deletion of neo and exon 6 leaving the lacZ
reporter gene intact. Mice totally lacking
DPP3 were bred by crossing mice
heterozygous for the mutant DPP3 allele
lacking neo and exon 6.
Serum and tissue lysate preparation
Animals were anesthetized with isoflurane and
blood was collected by the retro-orbital
puncture. Immediately following collection,
blood was allowed to clot and serum was
isolated by centrifugation. For tissue
collection, mice were sacrificed by cervical
dislocation and tissues were surgically
removed and washed with cold PBS.
Homogenization was performed on ice in
solution A (0.25M sucrose, 1 mM EDTA, 20
μM dithiothreitol, 0.1% Triton X-100, 20
μg/mL leupeptin, 2 μg/mL antipain, 1 μg/mL
pepstatin, pH 7.0) using an Ultra Turrax (IKA,
Staufen, Germany). 20,000 g infranatant was
used for further experiments. Protein
concentrations in the tissue lysates were
estimated using the Protein Assay Dye Reagent
(Bio-Rad, Munich, Germany) using bovine
serum albumin as standard. Serum and tissue
samples were stored at -80 °C until further
analysis.
SDS PAGE and Western blotting
Tissue lysates were diluted in Laemmli’s
sample buffer, and 20 µg of total protein/lane
at FACHBIBLIOTHEK CHEMIE on June 16, 2020http://www.jbc.org/Downloaded from
8
was subjected to sodium dodecyl sulfate-
polyacrylamide gel electrophoresis (SDS-
PAGE) using 10% SDS-polyacrylamide gels.
The resolved proteins were transferred onto
polyvinylidene difluoride (PVDF) membranes
(VWR, Pennsylvania, USA) using a Trans-
Blot SD transfer cell (Bio-Rad, CA, USA).
Following transfer, membranes were washed
with Tris-buffered saline containing 0.01%
Tween-20 (TBST) and then blocked in 5%
non-fat milk for 1 h at room temperature. The
membranes were then incubated overnight
with anti-DPP3 rabbit polyclonal antibody
(1:1,500, Proteintech Europe, Manchester, UK)
in TBST containing 5% non-fat milk at 4 ºC.
After washing three times for 10 min in TBST,
membranes were incubated with peroxidase-
labeled secondary antibody (1:5,000; Cell
Signaling Technology®, Danvers, MA, USA)
for 1 h at room temperature. The immunoblots
were developed using enhanced
chemiluminescent western blotting substrate
solution (Pierce-Thermo Fisher Scientific,
Waltham, MA, USA).
DPP3 activity assay in mouse tissue lysates
DPP3 activities in tissue lysates were
determined by fluorometrically (excitation,
332 nm; emission, 420 nm) measuring the
liberation of 2-naphthylamine at 37 °C in a
mixture containing 25 µl of 200 µM Arg-Arg-
2-naphthylamide as substrate in TBS buffer
(50 mM Tris, 100 mM NaCl, pH 8.2) and
tissue lysate equivalent to 20 µg of total
protein in a reaction mixture of 235 µl (White,
Tissue Culture treated Krystal 2000 96-well
plate from Porvair sciences, Norfolk, UK). The
activity assay was performed by continuous
measurement of fluorescence of 2-
naphthylamide for 30 min (Fluorescent plate
reader from Molecular Devices, Sunnyvale
CA, USA). The reaction was started by the
addition of the substrate. The samples were
measured in triplicates.
Body composition and metabolic
phenotyping
Lean and fat mass of mice were analyzed by
NMR (the minispec, NMR Analyzer, Bruker,
Ettlingen, Germany). To measure spontaneous
physical activity, O2 consumption, CO2
production, food and water intake, mice were
housed in metabolic cages allowing continuous
measurement of these parameters (LabMaster,
TSE Systems GmbH, Bad Homburg,
Germany). For measurements of energy
balance, animals were familiarized with these
cages for at least 72 hours before data
collection.
Analysis of angiotensin peptides in serum
Serum equilibration was performed at 37 °C
followed by stabilization of equilibrium
angiotensin levels and subsequent
quantification by LC-MS/MS analysis (50).
Briefly, stable isotope-labeled internal
standards for each Ang metabolite [Ang I (1-
10), Ang II (1-8), Ang (17), Ang (15), Ang
III (28), Ang IV (38), Ang (1-9), Ang (3-7),
Ang (2-7), Ang (2-10)] were added to
stabilized serum samples at a concentration of
200 pg/mL. Following C18-based solid-phase-
extraction, samples were subjected to LC-
MS/MS analysis using a reversed-phase
analytical column (Acquity UPLC® C18,
Waters, USA) operating in line with a XEVO
TQ-S triple quadrupole mass spectrometer
(Waters Xevo TQ/S, Milford, Massachusetts,
USA) in multiple reaction monitoring modes.
The internal standard was used to correct for
analyte recovery across the sample preparation
procedure in each sample. Analyte
concentrations were calculated from integrated
chromatograms considering the corresponding
response factors determined in appropriate
calibration curves in serum matrix.
Determination of ROS generation
The intracellular ROS level was detected by
using 2′,7′-dichlorodihydrofluorescein
diacetate (H2DCFDA) (Sigma Aldrich). When
oxidized by various active oxygen species, it is
irreversibly converted to the fluorescent form,
DCF (51, 52). ROS in kidney tissue was
estimated by diluting tissue lysate equivalent to
100 µg of total protein in ice-cold 40 mM Tris-
HCl buffer (pH 7.4). The samples were divided
into two equal fractions. In one fraction 40 µl
of 10 µM H2DCFDA in methanol was added
for ROS estimation. Another fraction with 40
µl of methanol was used as a control for tissue
auto-fluorescence. All samples were incubated
at 37 °C for 15 min and fluorescence was
determined at 485 nm excitation and 525 nm
emission using a fluorescence plate reader
(Molecular Devices, Sunnyvale, CA, USA). To
quantitate ROS levels, relative
at FACHBIBLIOTHEK CHEMIE on June 16, 2020http://www.jbc.org/Downloaded from
9
dichlorofluorescein fluorescence was used as a
standard.
Detection of lipid peroxidation activity
The extent of lipid peroxidation in kidney was
assessed using thiobarbituric acid reactive
substances (TBARS) as an index. For the
assay, kidney tissue equivalent to 1 mg of total
protein was incubated with 20% TCA and
0.67% TBA. The reaction mixture was heated
at 100 °C for 30 min and then cooled in an ice-
bath for 10 min. The samples were then
centrifuged at 3,000 rpm for 15 min. The
supernatant was collected to measure
absorbance at 532 nm. The formation of
TBARS was expressed using malondialdehyde
(MDA) equivalent as a standard.
Catalase activity
Catalase activity was measured as described in
(53). Briefly, kidney tissue equivalent to 1 mg
of total protein in 0.01 M PBS was incubated
with 0.2 M H2O2. The reaction was stopped by
adding 5% dichromate solution at 30 sec
intervals. The samples were heated at 60 °C for
10 min where the blue precipitate formed was
decomposed to a green solution. Consumption
of H2O2 was determined by recording
absorbance at 570 nm. A standard curve
containing 0 to 100 μM of H2O2 was prepared
to determine the amount of H2O2 present in
each sample.
Histological analysis of kidney
For the analysis of morphological differences,
kidneys were fixed in 4% neutral buffered
formaldehyde solution for 24 h, embedded in
paraffin, and further processed for periodic
acidSchiff (PAS) staining (2 µm thick
sections).
Blood pressure measurements
The CODA 8-channel non-invasive tail-cuff
technique (Kent Scientific, EMKA
Technologies, Paris, France) was used for
blood pressure measurements in young adult
male mice from 12-16 weeks of age. This
system uses Volume Pressure recording (VPR)
to detect blood pressure based on volume
changes in the tail (54). VPR cuffs were
checked routinely before the start of the
experiments. Heating pads were preheated to
35 °C before and during the measurements.
Each measurement consisted of 5 acclimation
cycles followed by 15 cycles during which the
systolic and diastolic pressure were measured.
Acclimation cycles were not used in blood
pressure analysis.
Isothermal titration calorimetry (ITC)
Calorimetric activity of six different
angiotensin peptides as purported substrates of
DPP3 were further assayed using a MicroCal
PEAQ-ITC (Malvern Panalytical Ltd,
Worcestershire, United Kingdom). The
peptides used for this assay, Ang I (1-10), Ang
II (1-8), Ang III (2-8), Ang IV (3-8), Ang (1-
5), and Ang (1-7), were commercially
purchased (Bachem, Bubendorf, Switzerland).
For this, the single injection method was used
where 5µl of 2 mM angiotensin peptide was
titrated into 200 µl of 20 µM purified hDPP3.
The purification of recombinant hDPP3 was
done as described in (35). Both the ligand and
the purified protein were prepared in 50 mM
Tris-HCl at pH 8.0 containing 100 mM NaCl.
The experiments consisted of a single injection
of the ligand continuously over a period of 10
seconds, including an initial delay of 180
seconds before the injection and a spacing of
300 seconds after the injection. The
experiments were performed at 25 °C with a
stirring speed of 500 rpm. The thermodynamic
characterization of this binding interaction was
obtained using MicroCal PEAQ-ITC Analysis
Software.
Statistical analysis
All data are expressed as mean ± standard
deviation (SD). Results were assessed using
two-tailed unpaired Student’s t-test (GraphPad
Prism 5, San Diego, USA). A P-value less than
0.05 was considered significant.
Data availability
All data presented and discussed are contained
within the article.
at FACHBIBLIOTHEK CHEMIE on June 16, 2020http://www.jbc.org/Downloaded from
10
Acknowledgements
This work was supported by a grant from the Austrian Science Foundation (FWF) through grants
W901 (Doctoral Program “Molecular Enzymology”) to KG, PM, and RZ. The authors are also
grateful for the support by the interuniversity program in natural sciences (NAWI Graz). We thank
Emilia Strandback, Karin Koch, and Chaitanya R. Tabib for experimental advice and discussions. We
also thank Margarete Lechleitner for her help in blood pressure measurements.
Conflict of interest
The authors declare that they have no conflicts of interest with the contents of this article.
References
1. Prajapati, S. C., and Chauhan, S. S. (2011) Dipeptidyl peptidase III: a multifaceted
oligopeptide N-end cutter. FEBS J. 278, 32563276
2. Sabljić, I., Meštrović, N., Vukelić, B., Macheroux, P., Gruber, K., Luić, M., and Abramić, M.
(2017) Crystal structure of dipeptidyl peptidase III from the human gut symbiont Bacteroides
thetaiotaomicron. PLoS One. 12, e0187295
3. Baral, P. K., Jajcanin-Jozić, N., Deller, S., Macheroux, P., Abramić, M., and Gruber, K. (2008)
The first structure of dipeptidyl-peptidase III provides insight into the catalytic mechanism and
mode of substrate binding. J. Biol. Chem. 283, 2231622324
4. Bezerra, G. A., Dobrovetsky, E., Viertlmayr, R., Dong, A., Binter, A., Abramić, M.,
Macheroux, P., Dhe-Paganon, S., and Gruber, K. (2012) Entropy-driven binding of opioid
peptides induces a large domain motion in human dipeptidyl peptidase III. Proc. Natl. Acad.
Sci. 109, 65256530
5. Kumar, P., Reithofer, V., Reisinger, M., Wallner, S., Pavkov-Keller, T., Macheroux, P., and
Gruber, K. (2016) Substrate complexes of human dipeptidyl peptidase III reveal the
mechanism of enzyme inhibition. Sci. Rep. 6, 23787
6. Chiba, T., Li, Y.-H., Yamane, T., Ogikubo, O., Fukuoka, M., Arai, R., Takahashi, S., Ohtsuka,
T., Ohkubo, I., and Matsui, N. (2003) Inhibition of recombinant dipeptidyl peptidase III by
synthetic hemorphin-like peptides. Peptides. 24, 773778
7. Sato, H., Kimura, K., Yamamoto, Y., and Hazato, T. (2003) [Activity of DPP III in human
cerebrospinal fluid derived from patients with pain]. Masui. 52, 257263
8. Cruz-Diaz, N., Wilson, B. A., and Chappell, M. C. (2017) Peptidases and the Renin-
Angiotensin System: The Alternative Angiotensin-(1-7) Cascade. Enzym. Inhib. Act.
10.5772/65949
9. Pang, X., Shimizu, A., Kurita, S., Zankov, D. P., Takeuchi, K., Yasuda-Yamahara, M., Kume,
S., Ishida, T., and Ogita, H. (2016) Novel Therapeutic Role for Dipeptidyl Peptidase III in the
Treatment of Hypertension. Hypertens. (Dallas, Tex. 1979). 68, 630641
10. Hast, B. E., Goldfarb, D., Mulvaney, K. M., Hast, M. A., Siesser, P. F., Yan, F., Hayes, D. N.,
and Major, M. B. (2013) Proteomic analysis of ubiquitin ligase KEAP1 reveals associated
proteins that inhibit NRF2 ubiquitination. Cancer Res. 73, 21992210
11. Menale, C., Robinson, L. J., Palagano, E., Rigoni, R., Erreni, M., Almarza, A. J., Strina, D.,
Mantero, S., Lizier, M., Forlino, A., Besio, R., Monari, M., Vezzoni, P., Cassani, B., Blair, H.
C., Villa, A., and Sobacchi, C. (2019) Absence of Dipeptidyl Peptidase 3 increases oxidative
at FACHBIBLIOTHEK CHEMIE on June 16, 2020http://www.jbc.org/Downloaded from
11
stress and causes bone loss. J. Bone Miner. Res. Off. J. Am. Soc. Bone Miner. Res.
10.1002/jbmr.3829
12. Brewster, U. C., Setaro, J. F., and Perazella, M. A. (2003) The renin-angiotensin-aldosterone
system: cardiorenal effects and implications for renal and cardiovascular disease states. Am. J.
Med. Sci. 326, 1524
13. Perazella, M. A., and Setaro, J. F. (2003) Renin-angiotensin-aldosterone system: fundamental
aspects and clinical implications in renal and cardiovascular disorders. J. Nucl. Cardiol. Off.
Publ. Am. Soc. Nucl. Cardiol. 10, 184196
14. Daniels, D., Mietlicki, E. G., Nowak, E. L., and Fluharty, S. J. (2008) Angiotensin II stimulates
water and NaCl intake through separate cell signalling pathways. Exp. Physiol. 94, 130137
15. Cat, A. N. D., Montezano, A. C., Burger, D., and Touyz, R. M. (2013) Angiotensin II, NADPH
oxidase, and redox signaling in the vasculature. Antioxid. Redox Signal. 19, 11101120
16. Hitomi, H., Kiyomoto, H., and Nishiyama, A. (2007) Angiotensin II and oxidative stress. Curr.
Opin. Cardiol. 22, 311
17. Crowley, S. D., Gurley, S. B., Herrera, M. J., Ruiz, P., Griffiths, R., Kumar, A. P., Kim, H.-S.,
Smithies, O., Le, T. H., and Coffman, T. M. (2006) Angiotensin II causes hypertension and
cardiac hypertrophy through its receptors in the kidney. Proc. Natl. Acad. Sci. U. S. A. 103,
1798517990
18. Sachse, A., and Wolf, G. (2007) Angiotensin II-induced reactive oxygen species and the
kidney. J. Am. Soc. Nephrol. 18, 24392446
19. de Cavanagh, E. M. V, Inserra, F., Ferder, M., and Ferder, L. (2007) From Mitochondria to
Disease: Role of the Renin-Angiotensin System. Am. J. Nephrol. 27, 545553
20. Tschöp, M. H., Speakman, J. R., Arch, J. R. S., Auwerx, J., Brüning, J. C., Chan, L., Eckel, R.
H., Farese, R. V., Galgani, J. E., Hambly, C., Herman, M. A., Horvath, T. L., Kahn, B. B.,
Kozma, S. C., Maratos-Flier, E., Müller, T. D., Münzberg, H., Pfluger, P. T., Plum, L.,
Reitman, M. L., Rahmouni, K., Shulman, G. I., Thomas, G., Kahn, C. R., and Ravussin, E.
(2012) A guide to analysis of mouse energy metabolism. Nat. Methods. 9, 5763
21. Cruz-Diaz, N., Wilson, B. A., Pirro, N. T., Brosnihan, K. B., Marshall, A. C., and Chappell, M.
C. (2016) Identification of dipeptidyl peptidase 3 as the Angiotensin-(17) degrading peptidase
in human HK-2 renal epithelial cells. Peptides. 83, 2937
22. Kihara, M., Umemura, S., Sumida, Y., Yokoyama, N., Yabana, M., Nyui, N., Tamura, K.,
Murakami, K., Fukamizu, A., and Ishii, M. (1998) Genetic deficiency of angiotensinogen
produces an impaired urine concentrating ability in mice. Kidney Int. 53, 548555
23. Klein, J. D., Quach, D. Le, Cole, J. M., Disher, K., Mongiu, A. K., Wang, X., Bernstein, K. E.,
and Sands, J. M. (2002) Impaired urine concentration and absence of tissue ACE: involvement
of medullary transport proteins. Am. J. Physiol. Renal Physiol. 283, 517
24. Kim, S.-M., Kim, Y.-G., Jeong, K.-H., Lee, S.-H., Lee, T.-W., Ihm, C.-G., and Moon, J.-Y.
(2012) Angiotensin II-Induced Mitochondrial Nox4 Is a Major Endogenous Source of
Oxidative Stress in Kidney Tubular Cells. PLoS One. 7, e39739
25. Fitzsimons, J. T. (1998) Angiotensin, thirst, and sodium appetite. Physiol. Rev. 78, 583686
26. Lazartigues, E., Sinnayah, P., Augoyard, G., Gharib, C., Johnson, A. K., and Davisson, R. L.
(2008) Enhanced water and salt intake in transgenic mice with brain-restricted overexpression
of angiotensin (AT1) receptors. Am. J. Physiol. - Regul. Integr. Comp. Physiol. 295, 1539
at FACHBIBLIOTHEK CHEMIE on June 16, 2020http://www.jbc.org/Downloaded from
12
1545
27. Benigni, A., Cassis, P., and Remuzzi, G. (2010) Angiotensin II revisited: new roles in
inflammation, immunology and aging. EMBO Mol. Med. 2, 247257
28. Wen, H., Gwathmey, J. K., and Xie, L.-H. (2012) Oxidative stress-mediated effects of
angiotensin II in the cardiovascular system. World J. Hypertens. 2, 34
29. Lu, K., Alcivar, A. L., Ma, J., Foo, T. K., Zywea, S., Mahdi, A., Huo, Y., Kensler, T. W.,
Gatza, M. L., and Xia, B. (2017) NRF2 Induction Supporting Breast Cancer Cell Survival Is
Enabled by Oxidative Stress-Induced DPP3-KEAP1 Interaction. Cancer Res. 77, 28812892
30. Zhang, D. D. (2006) Mechanistic studies of the Nrf2-Keap1 signaling pathway. Drug Metab.
Rev. 38, 769789
31. Jaramillo, M. C., and Zhang, D. D. (2013) The emerging role of the Nrf2-Keap1 signaling
pathway in cancer. Genes Dev. 27, 21792191
32. Kansanen, E., Kuosmanen, S. M., Leinonen, H., and Levonen, A.-L. (2013) The Keap1-Nrf2
pathway: Mechanisms of activation and dysregulation in cancer. Redox Biol. 1, 4549
33. Deshmukh, P., Unni, S., Krishnappa, G., and Padmanabhan, B. (2017) The Keap1-Nrf2
pathway: promising therapeutic target to counteract ROS-mediated damage in cancers and
neurodegenerative diseases. Biophys. Rev. 9, 4156
34. Liu, Y., Kern, J. T., Walker, J. R., Johnson, J. A., Schultz, P. G., and Luesch, H. (2007) A
genomic screen for activators of the antioxidant response element. Proc. Natl. Acad. Sci. U. S.
A. 104, 52055210
35. Rehfeld, L., Funk, E., Jha, S., Macheroux, P., Melander, O., and Bergmann, A. (2018) Novel
Methods for the Quantification of Dipeptidyl Peptidase 3 (DPP3) Concentration and Activity in
Human Blood Samples. J. Appl. Lab. Med. 10.1373/jalm.2018.027995
36. Takagi, K., Blet, A., Levy, B., Deniau, B., Azibani, F., Feliot, E., Bergmann, A., Santos, K.,
Hartmann, O., Gayat, E., Mebazaa, A., and Kimmoun, A. (2020) Circulating dipeptidyl
peptidase 3 and alteration in haemodynamics in cardiogenic shock: results from the OptimaCC
trial. Eur. J. Heart Fail. 22, 279286
37. Deniau, B., Rehfeld, L., Santos, K., Dienelt, A., Azibani, F., Sadoune, M., Kounde, P. R.,
Samuel, J. L., Tolpannen, H., Lassus, J., Harjola, V.-P., Vodovar, N., Bergmann, A.,
Hartmann, O., Mebazaa, A., and Blet, A. (2020) Circulating dipeptidyl peptidase 3 is a
myocardial depressant factor: dipeptidyl peptidase 3 inhibition rapidly and sustainably
improves haemodynamics. Eur. J. Heart Fail. 22, 290299
38. Dépret, F., Amzallag, J., Pollina, A., Fayolle-Pivot, L., Coutrot, M., Chaussard, M., Santos, K.,
Hartmann, O., Jully, M., Fratani, A., Oueslati, H., Cupaciu, A., Benyamina, M., Guillemet, L.,
Deniau, B., Mebazaa, A., Gayat, E., Farny, B., Textoris, J., and Legrand, M. (2020) Circulating
dipeptidyl peptidase-3 at admission is associated with circulatory failure, acute kidney injury
and death in severely ill burn patients. 10.1186/s13054-020-02888-5
39. Yu, L., Yuan, K., Phuong, H. T. A., Park, B. M., and Kim, S. H. (2016) Angiotensin-(1-5), an
active mediator of renin-angiotensin system, stimulates ANP secretion via Mas receptor.
Peptides. 86, 3341
40. de Bold, A. J., Borenstein, H. B., Veress, A. T., and Sonnenberg, H. (1981) A rapid and potent
natriuretic response to intravenous injection of atrial myocardial extract in rats. Life Sci. 28,
8994
at FACHBIBLIOTHEK CHEMIE on June 16, 2020http://www.jbc.org/Downloaded from
13
41. Shono, M., Yoshimura, M., Nakayama, M., Yamamuro, M., Abe, K., Suzuki, S., Mizuno, Y.,
Sugiyama, S., Saito, Y., Nakao, K., Yasue, H., and Ogawa, H. (2007) Predominant effect of A-
type natriuretic peptide on reduction of oxidative stress during the treatment of patients with
heart failure. Circ. J. 71, 10401046
42. Komukai, K., Mochizuki, S., and Yoshimura, M. (2010) Gender and the renin-angiotensin-
aldosterone system. Fundam. Clin. Pharmacol. 24, 687698
43. Toering, T. J., Van Der Graaf, A. M., Visser, F. W., Buikema, H., Navis, G., Faas, M. M., and
Lely, A. T. (2015) Gender differences in response to acute and chronic angiotensin II infusion:
A translational approach. Physiol. Rep. 3, 111
44. Tatchum-Talom, R., Eyster, K. M., and Martin, D. S. (2005) Sexual dimorphism in angiotensin
II-induced hypertension and vascular alterations. Can. J. Physiol. Pharmacol. 83, 413422
45. Xue, B., Pamidimukkala, J., and Hay, M. (2005) Sex differences in the development of
angiotensin II-induced hypertension in conscious mice. Am. J. Physiol. Heart Circ. Physiol.
288, H2177H2184
46. Xue, B., Pamidimukkala, J., Lubahn, D. B., and Hay, M. (2007) Estrogen receptor-alpha
mediates estrogen protection from angiotensin II-induced hypertension in conscious female
mice. Am. J. Physiol. Heart Circ. Physiol. 292, H1770H1776
47. Gupte, M., Thatcher, S. E., Boustany-Kari, C. M., Shoemaker, R., Yiannikouris, F., Zhang, X.,
Karounos, M., and Cassis, L. A. (2012) Angiotensin converting enzyme 2 contributes to sex
differences in the development of obesity hypertension in C57BL/6 mice. Arterioscler.
Thromb. Vasc. Biol. 32, 13921399
48. Mačak Šafranko, Sobočanec, S., Šarić, A., Jajčanin-Jozić, N., Krsnik, Aralica, G., Balog, T.,
and Abramić, M. (2015) The effect of 17β-estradiol on the expression of dipeptidyl peptidase
III and heme oxygenase 1 in liver of CBA/H mice. J. Endocrinol. Invest. 38, 471479
49. Sobočanec, S., Filić, V., Matovina, M., Majhen, D., Šafranko, Ž. M., Hadžija, M. P., Krsnik,
Ž., Kurilj, A. G., Šarić, A., Abramić, M., and Balog, T. (2016) Prominent role of exopeptidase
DPP III in estrogen-mediated protection against hyperoxia in vivo. Redox Biol. 8, 149159
50. Sharp, S., Poglitsch, M., Zilla, P., Davies, N. H., and Sturrock, E. D. (2015) Pharmacodynamic
effects of C-domain-specific ACE inhibitors on the renin-angiotensin system in myocardial
infarcted rats. J. renin-angiotensin-aldosterone Syst. JRAAS. 16, 11491158
51. Pavelescu, L. A. (2015) On reactive oxygen species measurement in living systems. J. Med.
Life. 8, 3842
52. Dikalov, S., Griendling, K. K., and Harrison, D. G. (2007) Measurement of Reactive Oxygen
Species in Cardiovascular Studies. Hypertension. 49, 717727
53. Sinha, A. K. (1972) Colorimetric assay of catalase. Anal. Biochem. 47, 389394
54. Wilde, E., Aubdool, A. A., Thakore, P., Baldissera, L., Alawi, K. M., Keeble, J., Nandi, M.,
and Brain, S. D. (2017) Tail-Cuff Technique and Its Influence on Central Blood Pressure in the
Mouse. J. Am. Heart Assoc. 10.1161/JAHA.116.005204
at FACHBIBLIOTHEK CHEMIE on June 16, 2020http://www.jbc.org/Downloaded from
14
Footnotes
The abbreviations used are:
DPP3, dipeptidyl peptidase 3; DPP3+/+, wild-type; DPP3-/-, DPP3-knockout; RAS, renin-angiotensin
system; Ang, angiotensin; TCA, tricarboxylic acid; ROS, reactive oxygen species; LC-MS/MS, liquid
chromatography-tandem mass spectrometry; ITC, isothermal titration calorimetry; PCA, principal
component analysis; OPLS-DA, orthogonal projections to latent structures discriminant analysis;
Sirt3, sirtuin-3; PARP, poly (ADP-ribose) polymerase; ACE, angiotensin converting enzyme; NEP,
neutral endopeptidase; PEP, prolyl endopeptidase; PCP, prolyl carboxypeptidase
at FACHBIBLIOTHEK CHEMIE on June 16, 2020http://www.jbc.org/Downloaded from
15
Tables
Table 1: Kinetic parameters obtained from single injection ITC experiments with hDPP3 and
angiotensin peptides. The Michaelis-Menten constant KM and maximum turnover number kcat are
obtained from the fit to a MichaelisMenten curve for the reaction rate as a function of substrate
concentration. The enthalpy of interaction (ΔH) (also called heat of reaction) is a direct measurement
of the rate at which heat is exchanged with the surroundings. Data represents mean ± SD. All data
points are 3 or more technical replicates from 2 biological replicates.
Kinetic, calorimetric parameters of hDPP3 and angiotensin interaction
ΔH (kcal/mol)
kcat (s-1)
Ang II
-1.97 ± -0.06
0.25 ± 0.007
Ang (1-7)
-1.52 ± -0.10
0.14 ± 0.003
Ang (1-5)
-1.84 ± -0.04
0.35 ± 0.002
at FACHBIBLIOTHEK CHEMIE on June 16, 2020http://www.jbc.org/Downloaded from
16
Figures
Figure 1. Generation and validation of DPP3 knockout mice. (A), Strategy for the generation of
DPP3-knockout mice. B-C, Demonstration of the lack of DPP3 in male DPP3-/- mice by Western blot
(B) and activity assays using Arg-Arg-2-naphthylamide as an artificial substrate (C) (BAT = brown
adipose tissue; SI = small intestine; SM = skeletal muscle; WAT = white adipose tissue) (12-16 weeks
of age; n= 3/group). n.d.= non detectable, **p<0.01, ***p<0.001 versus wild-type mice based on
unpaired two-sided Student’s t-test. Data is representative of 3 technical replicates from 3 biological
replicates and presented as mean ± SD.
at FACHBIBLIOTHEK CHEMIE on June 16, 2020http://www.jbc.org/Downloaded from
17
Figure 2. DPP3-/- mice display lower body weight and altered food and water intake. (A) Body
weight and (B) body composition of male mice fed a regular chow diet. C-H, Cumulative (top panel)
and total (bottom panel) food intake (C and F, respectively), water consumption (D and G,
respectively) and locomotive motion (E and H, respectively), were measured in metabolic cages over
the light and dark phases in male DPP3-/- and DPP3+/+ mice (12-16 weeks of age; n=6/group) fed a
regular chow diet over a period of six consecutive days. *p<0.05, **p<0.01 versus wild-type mice
based on unpaired two-sided Student’s t-test. Data are representative for two independent cohorts and
presented as mean ± SD.
at FACHBIBLIOTHEK CHEMIE on June 16, 2020http://www.jbc.org/Downloaded from
18
Figure 3. DPP3 knockout mice display unaltered energy expenditure. Cumulative and daily
oxygen consumption (A and D respectively) and carbon-dioxide production (B and E respectively),
RER (C) and EE (F) were measured in metabolic cages over the light and dark phases in male DPP3-/-
and DPP3+/+ mice (12-16 weeks of age; n=6/group) fed a regular chow diet over a period of six
consecutive days. *p<0.05, **p<0.01 versus wild-type mice based on unpaired two-sided Student’s t-
test. Data are representative for two independent cohorts and presented as mean ± SD.
at FACHBIBLIOTHEK CHEMIE on June 16, 2020http://www.jbc.org/Downloaded from
19
Figure 4. DPP3-/- mice exhibit significantly elevated water consumption during fasting. (A)
Cumulative and (B) daily water intake during a 13h fasting period was measured in metabolic cages in
male DPP3-/- and DPP3+/+ mice (12-16 weeks of age; n=6/group). *p<0.05, **p<0.01 versus wild-type
mice based on unpaired two-sided Student’s t-test. Data represents mean ± SD.
at FACHBIBLIOTHEK CHEMIE on June 16, 2020http://www.jbc.org/Downloaded from
20
Figure 5. DPP3-/- mice exhibit increased circulating angiotensin metabolites. (A) Concentration of
RAS peptides, (B) aldosterone, and (C) renin activity in serum measured by liquid chromatography-
mass spectrometry in male DPP3-/- and DPP3+/+ mice (12-16 weeks of age; n = 8/group). *p<0.05,
**p<0.01, ***p<0.001 versus wild-type mice based on unpaired two-sided Student’s t-test. Data
represents mean + SD.
at FACHBIBLIOTHEK CHEMIE on June 16, 2020http://www.jbc.org/Downloaded from
21
Figure 6. DPP3 knockout renders mice susceptible to oxidative stress. Quantification of (A) ROS
production, (B) Lipid peroxidation and (C) catalase activity in kidney homogenates of male DPP3-/-
and DPP3+/+ mice (12-16 weeks of age; n = 5/group). *p<0.05, **p<0.01 versus wild-type mice based
on unpaired two-sided Student’s t-test. Experiments were performed in technical triplicates and
represent mean + SD.
at FACHBIBLIOTHEK CHEMIE on June 16, 2020http://www.jbc.org/Downloaded from
22
Figure 7. DPP3 deletion does not affect morphology of mouse kidney. PAS (periodic acid Schiff) -
stained slides of renal histology showing unremarkable glomeruli and tubuli in (A) DPP3-/- and (B)
DPP3+/+ mice. Left 10x, right 20x.
at FACHBIBLIOTHEK CHEMIE on June 16, 2020http://www.jbc.org/Downloaded from
23
Figure 8. DPP3 deficiency does not lead to changes in blood pressure. Systolic and diastolic blood
pressure were measured by tail-cuff method in male DPP3+/+ and DPP3-/- mice (18-22 weeks of age; n
= 8/group). Data are presented as mean + SD.
at FACHBIBLIOTHEK CHEMIE on June 16, 2020http://www.jbc.org/Downloaded from
24
Figure 9. RAS peptides can be “good” or “bad” substrates of DPP3. RAS peptides turned over by
DPP3, thus acting as “good” substrates (A-C). Raw data showing the heat change of the reaction as a
function of time (top panel) and fitted curve for the rate of reaction (bottom panel) of (A) Ang II, (B)
Ang 1-7, and (C) Ang 1-5. Some RAS peptides demonstrated both endothermic and exothermic
behavior, thus acting as “slow” substrates of DPP3 (D-F). The biphasic peaks were most likely due to
binding to DPP3 and a subsequent slow turnover event. Raw data showing the heat change of the
reaction in the case of slow substrates, (D) tynorphin, (E) Ang III, and (F) Ang IV. Curve fitting was
not possible in the case of slow substrates. The reaction was started by injecting 5µl, 2 mM
angiotensin peptides to the calorimetric cell containing 20 µM hDPP3. Data represents 3 or more
technical replicates from 2 biological replicates.
at FACHBIBLIOTHEK CHEMIE on June 16, 2020http://www.jbc.org/Downloaded from
25
Figure 10. DPP3 exerts a sex-specific effect on the knockout mice. (A) Body weight and (B) body
composition of female mice fed a regular chow diet. (C) Daily food intake and (D) water consumption
was measured in metabolic cages over the light and dark phases in DPP3-/- and DPP3+/+ female mice
(12-16 weeks of age; n=6/group) fed a regular chow diet over a period of six consecutive days. (E)
Concentration of RAS peptides in serum measured by liquid chromatography-mass spectrometry in
DPP3-/- and DPP3+/+ female mice (12-16 weeks of age; n=8/group) and (F) Quantification of ROS
production in DPP3-/- and DPP3+/+ female mice (12-16 weeks of age; n = 5/group). *p<0.05, **p<0.01
versus wild-type mice based on unpaired two-sided Student’s t-test. Data represents mean ± SD.
at FACHBIBLIOTHEK CHEMIE on June 16, 2020http://www.jbc.org/Downloaded from
Gruber, Robert Zimmermann and Peter Macheroux
Marion Pollheimer, Lisa M. Pusch, Grazia Malovan, Sasa Frank, Tobias Madl, Karl
Shalinee Jha, Ulrike Taschler, Oliver Domenig, Marko Poglitsch, Benjamin Bourgeois,
Dipeptidyl peptidase 3 modulates the renin-angiotensin system in mice
published online June 16, 2020J. Biol. Chem.
10.1074/jbc.RA120.014183Access the most updated version of this article at doi:
Alerts:
When a correction for this article is posted When this article is cited
to choose from all of JBC's e-mail alertsClick here
at FACHBIBLIOTHEK CHEMIE on June 16, 2020http://www.jbc.org/Downloaded from
... The main functions observed so far in the alternative RAS are its ability to degrade Ang II via the activity of angiotensinconverting enzyme 2 (ACE2). The classical RAS can also be modulated through the action of peptidases, such as dipeptidyl peptidase 3 (DPP3), which can degrade peptides from both the classical and alternative RAS [14,15]. ...
... Despite its primarily intracellular location, circulating DPP3 has been detected among healthy blood donors [43]. It is hypothesized to be released during a cell death process in conditions as shock, where it is responsible for the cleavage of various peptides, including enkephalins, endorphins, and angiotensin peptides that are less than 10 residues in length such as Ang II, Ang-(1-7), or Ang-(1-5) [15]. ...
... DPP3 can be released into the circulation and modulate both the classical and alternative RAS, contributing to its imbalance [15,63]. In a prospective observational international study involving 585 patients with sepsis, high admission DPP3 levels were associated with an increased incidence of AKI within 7 days, greater use of renal replacement therapy (RRT), longer ICU stay, and higher mortality rates [64]. ...
Article
Full-text available
The renin–angiotensin system (RAS) plays a crucial role in regulating blood pressure and the cardio-renal system. The classical RAS, mainly mediated by angiotensin I, angiotensin-converting enzyme, and angiotensin II, has been reported to be altered in critically ill patients, such as those in vasodilatory shock. However, recent research has highlighted the role of some components of the counterregulatory axis of the classical RAS, termed the alternative RAS, such as angiotensin-converting Enzyme 2 (ACE2) and angiotensin-(1–7), or peptidases which can modulate the RAS like dipeptidyl-peptidase 3, in many critical situations. In cases of shock, dipeptidyl-peptidase 3, an enzyme involved in the degradation of angiotensin and opioid peptides, has been associated with acute kidney injury and mortality and preclinical studies have tested its neutralization. Angiotensin-(1–7) has been shown to prevent septic shock development and improve outcomes in experimental models of sepsis. In the context of experimental acute lung injury, ACE2 activity has demonstrated a protective role, and its inactivation has been associated with worsened lung function, leading to the use of active recombinant human ACE2, in preclinical and human studies. Angiotensin-(1–7) has been tested in experimental models of acute lung injury and in a recent randomized controlled trial for patients with COVID-19 related hypoxemia. Overall, the alternative RAS appears to have a role in the pathogenesis of disease in critically ill patients, and modulation of the alternative RAS may improve outcomes. Here, we review the available evidence regarding the methods of analysis of the RAS, pathophysiological disturbances of this system, and discuss how therapeutic manipulation may improve outcomes in the critically ill.
... DPP3 is an intracellular aminopeptidase that is ubiquitously expressed and highly conserved in higher animals where it is implicated in antioxidant response. When released into the bloodstream, circulating DPP3 (cDPP3) cleaves various peptides of the RAS, including Ang II [14,15]. It has been hypothesized that DPP3 is released into the circulation during sepsis, as a result of tissue injury and/or cell death [16,17], potentially leading to Ang II degradation and further increase in the Ang I/Ang II ratio [12,18,19]. ...
Article
Full-text available
Background Dipeptidyl peptidase 3 (DPP3) is a ubiquitous cytosolic enzyme released into the bloodstream after tissue injury, that can degrade angiotensin II. High concentrations of circulating DPP3 (cDPP3) have been associated with worse outcomes during sepsis. The aim of this study was to assess the effect of Procizumab (PCZ), a monoclonal antibody that neutralizes cDPP3, in an experimental model of septic shock. Methods In this randomized, open-label, controlled study, 16 anesthetized and mechanically ventilated pigs with peritonitis were randomized to receive PCZ or standard treatment when the mean arterial pressure (MAP) dropped below 50 mmHg. Resuscitation with fluids, antimicrobial therapy, peritoneal lavage, and norepinephrine was initiated one hour later to maintain MAP between 65–75 mmHg for 12 h. Hemodynamic variables, tissue oxygenation indices, and measures of organ failure and myocardial injury were collected. Organ blood flow was assessed using isotopic assessment ( 99m technetium albumin). cDPP3 activity, equilibrium analysis of the renin–angiotensin system and circulating catecholamines were measured. Tissue mRNA expression of interleukin-6 and downregulation of adrenergic and angiotensin receptors were assessed on vascular and myocardial samples. Results PCZ-treated animals had reduced cDPP3 levels and required less norepinephrine and fluid than septic control animals for similar organ perfusion and regional blood flow. PCZ-treated animals had less myocardial injury, and higher PaO 2 /FiO 2 ratios. PCZ was associated with lower circulating catecholamine levels; higher circulating angiotensin II and higher angiotensin II receptor type 1 myocardial protein expression, and with lower myocardial and radial artery mRNA interleukin-6 expression. Conclusions In an experimental model of septic shock, PCZ administration was associated with reduced fluid and catecholamine requirements, less myocardial injury and cardiovascular inflammation, along with preserved angiotensin II signaling. Graphical Abstract
... Ang IV binds to Ang II type IV receptor to exert vasodilation, cardioprotective effects, and natriuresis [14]. DPP3 does not cleave larger peptides (≥ 10 amino acids) such as Ang I [9,13,15,16]. ...
Article
Full-text available
The renin-angiotensin system (RAS) constitutes one of the principal mechanisms to maintain hemodynamic and fluid homeostasis. However, most research until now on RAS primarily focuses on its relationship with hypertension and its role in critically ill hypotensive populations is not well understood. With the approval of angiotensin II (Ang II) in the United States and Europe, following a phase 3 randomized controlled trial showing efficacy in catecholamine-resistant vasodilatory shock, there is growing interest in RAS in critically ill patients. Among the fundamental components of RAS, renin acts as the initial stimulus for the entire system. In the context of hypotension, its release increases in response to low blood pressure sensed by renal baroreceptors and attenuated negative Ang II feedback loop. Thus, elevated renin could reflect disease severity and predict poor outcomes. Studies investigating this hypothesis have validated the prognostic accuracy of renin in various critically ill populations, with several reports indicating its superiority to lactate for mortality prediction. Accordingly, renin reduction has been used to assess the effectiveness of Ang II administration. Furthermore, renin holds potential to identify patients who might benefit from Ang II treatment, potentially paving the way for personalized vasopressor management. Despite these promising data, most available evidence is derived from retrospective analysis and necessitates prospective confirmation. The absence of a rapid, point-of-care and reliable renin assay presents another hurdle to its integration into routine clinical practice. This narrative review aims to describe the current understanding and future directions of renin as a biomarker during resuscitation of critically ill patients.
... DPP3 is widely distributed in various tissues, such as red blood cells, white blood cells, lungs, heart, kidney, intestine, skeletal muscle, skin, brain, liver, and spleen, and partially in soluble form in circulating blood [2]. DPP3 can lyse a variety of bioactive peptides, including angiotensin, bilinogen enterohepatic circulation, enkephalin, endorphin and dipeptidyl derivatives, and is involved in a variety of pathophysiological processes, including blood pressure regulation, inflammation regulation and pain signaling [3,4]. In recent years, emerging studies have revealed that DPP3 is upregulated in aggressive brain glioma [5], ovarian cancer [6], endometrial cancer [7], and bowel cancer [8]. ...
Article
DPP3, a dipeptidyl peptidase, participates in a variety of pathophysiological processes. DPP3 is upregulated in cancer and might serve as a key factor in the tumorigenesis and progression of various malignancies. However, its specific role and molecular mechanism are still unknown. In this study, the expression of DPP3 in breast cancer tissues is analyzed using TCGA database. Kaplan-Meier survival analysis is performed to estimate the effect of DPP3 on the survival outcomes. To explore the biological function and mechanisms of DPP3 in breast cancer, biochemical and cell biology assays are conducted in vitro. DPP3 expresses at a higher level in breast cancer tissues than that in adjacent tissues in both TCGA database and clinical samples. Patients with high expression of DPP3 have poor survival outcomes. The proliferation and migration abilities of tumor cells with stable DPP3 knockout in breast cancer cell lines are significantly inhibited, and apoptosis is increased in vitro. GSEA analysis shows that DPP3 can affect lipid metabolism and fatty acid synthesis in tumors. Subsequent experiments show that DPP3 could stabilize FASN expression and thus promote fatty acid synthesis in tumor cells. The results of the metabolomic analysis also confirm that DPP3 can affect the content of free fatty acids. This study demonstrates that DPP3 plays a role in the reprogramming of fatty acid metabolism in tumors and is associated with poor prognosis in breast cancer patients. These findings will provide a new therapeutic target for the treatment of breast cancer.
... All peptides showed dipeptidyl peptidase (DPP)-III-and/or IV-inhibitory activity. DPP-III plays a role in blood pressure regulation by modulating angiotensin I and II [44]. Human DPP-IV is involved in blood glucose regulation, and its inhibition is used as a target for antidiabetic drugs [45]. ...
Article
Full-text available
The aim of the present study was to determine the ACE inhibitory activity of aqueous extracts of olive pomace and to understand whether they represent a good source of bioactive LMW peptides for nutritional and pharmacological applications. We produced a water extract from olive pomace (var. Picual) and obtained its low molecular weight (LMW) fraction (<3 kDa). The calculated yield of extraction was 100.2 ± 7.9 mg of LMW peptides per 100 g of olive pomace. The olive pomace LMW fraction possessed strong ACE inhibitory activity (IC50 = 3.57 ± 0.22 µg prot/mL). The LMW fraction (<3 kDa) was analysed by nanoscale liquid chromatography-Orbitrap coupled with tandem mass spectrometry and de novo sequencing. Thirty new peptides, containing between 7–17 amino acids and molecular masses ranging 778–1354 Da, were identified by the Peaks database algorithm using the available Olea europaea (cv. Farga) genome database. Ten new peptides were also identified by Peaks de novo sequencing. The protein sources of twelve peptides detected in the database by Peaks DB were identified by BLAST search. The ACE inhibitory activity of the identified peptides was predicted by BIOPEP software. We conclude that olive pomace possesses ACE inhibitory activity and contains low molecular weight peptides with (predicted) biological activity. Olive pomace may represent a good source of peptides for nutritional and pharmaceutical applications. In our study, it has been shown that olive pomace possesses ACE inhibitory activity and contains low molecular weight peptides with (predicted) biological activity. Olive pomace may represent a good source of peptides for nutritional and pharmaceutical applications. More research is needed in order to identify the in vivo effects of olive pomace bioactive peptides.
... Another candidate biomarker to guide the use of AT2 is dipeptidyl peptidase 3 (DPP3), an aminopeptidase that cleaves a variety of biologically active oligopeptides including AT2 [17][18][19]. In animal models, DPP3 modulates the renin-angiotensin system by cleaving AT2 without acting on angiotensin I (AT1), leading to an elevated AT1/AT2 ratio [20]. Elevated AT1/AT2 ratios were observed in patients with catecholamine-resistant septic shock in ATHOS-3, and, like elevated renin, an elevated AT1/AT2 ratio may identify patients likely to benefit from AT2 therapy [21]. ...
Article
Full-text available
Background Data to support the use of specific vasopressors in septic shock are limited. Since angiotensin II (AT2) was approved by the Food and Drug Administration in 2017, multiple mechanistically distinct vasopressors are available to treat septic shock, but minimal data exist regarding which patients are most likely to benefit from each agent. Renin and dipeptidyl peptidase 3 (DPP3) are components of the renin–angiotensin–aldosterone system which have been shown to outperform lactate in predicting sepsis prognosis, and preliminary data suggest they could prove useful as biomarkers to guide AT2 use in septic shock. Methods The DARK-Sepsis trial is an investigator-initiated industry-funded, open-label, single-center randomized controlled trial of the use of AT2 versus standard of care (SOC) vasopressor therapy in patients admitted to the intensive care unit (ICU) with vasodilatory shock requiring norepinephrine ≥ 0.1 mcg/kg/min. In both groups, a series of renin and DPP3 levels will be obtained over the first 24 h of treatment with AT2 or SOC. The primary study outcome will be the ability of these biomarkers to predict response to vasopressor therapy, as measured by change in total norepinephrine equivalent dose of vasopressors at 3 h post-drug initiation or the equivalent timepoint in the SOC arm. To determine if the ability to predict vasopressor response is specific to AT2 therapy, the primary analysis will be the ability of baseline renin and DPP3 levels to predict vasopressor response adjusted for treatment arm (AT2 versus control) and Sequential Organ Failure Assessment (SOFA) scores. Secondary outcomes will include rates of acute kidney injury, need for mechanical ventilation and kidney replacement therapy, lengths of stay in the ICU and hospital, ICU and hospital mortality, and rates of prespecified adverse events. Discussion With an armamentarium of mechanistically distinct vasopressor agents now available, sub-phenotyping patients using biomarkers has the potential to improve septic shock outcomes by enabling treatment of the correct patient with the correct vasopressor at the correct time. However, this approach requires validation in a large definitive multicenter trial. The data generated through the DARK-Sepsis study will prove crucial to the optimal design and patient enrichment of such a pivotal trial. Trial registration ClinicalTrials.gov NCT05824767. Registered on April 24, 2023.
... Most of its substrates have been identified in ex vivo or in vitro experiments and include angiotensin peptides and endogenous opioids such as enkephalins and endomorphins. [7][8][9] Low cDPP3 concentration in the bloodstream was found in healthy animals and individuals and its role in the metabolism of its known substrates remains poorly understood. Interestingly, there is a strong correlation between cDPP3 concentration and activity in critically ill patients confirming that, in acute conditions, DPP3 (dipeptidyl peptidase 3) is released in its active form. ...
Article
BACKGROUND High circulating DPP3 (dipeptidyl peptidase 3) has been associated with poor prognosis in critically ill patients with circulatory failure. In such situation, DPP3 could play a pathological role, putatively via an excessive angiotensin peptides cleavage. Our objective was to investigate the hemodynamics changes induced by DPP3 in mice and the relation between the observed effects and renin–angiotensin system modulation. Methods Ten-week-old male C57Bl/6J mice were subjected to intravenous injection of purified human DPP3 or an anti-DPP3 antibody (procizumab). Invasive blood pressure and renal blood flow were monitored throughout the experiments. Circulating angiotensin peptides and catecholamines were measured and receptor blocking experiment performed to investigate the underlying mechanisms. RESULTS DPP3 administration significantly increased renal blood flow, while blood pressure was minimally affected. Conversely, procizumab led to significantly decreased renal blood flow. Angiotensin peptides measurement and an AT1R (angiotensin II receptor type 1) blockade experiment using valsartan demonstrated that the renovascular effect induced by DPP3 is due to reduced AT1R activation via decreased concentrations of circulating angiotensin II, III, and IV. Measurements of circulating catecholamines and an adrenergic receptor blockade by labetalol demonstrated a concomitant catecholamines release that explains blood pressure maintenance upon DPP3 administration. CONCLUSIONS High circulating DPP3 increases renal blood flow due to reduced AT1R activation via decreased concentrations of circulating angiotensin peptides while blood pressure is maintained by concomitant endogenous catecholamines release.
... It is known to cleave various peptides, including angiotensins, enkephalins, and endorphins. Notably, DPP3 has been identified as cleaving various peptides of the RAS, including Ang II, Ang-(1-7), and Angiotensin-(1-5) [40]. During sepsis, DPP3 is hypothesized to be released into the circulation due to a cell death process [39]. ...
Article
Purpose of review This review aims to explore the relationship between the renin angiotensin system (RAS) and sepsis-associated acute kidney injury (SA-AKI), a common complication in critically ill patients associated with mortality, morbidity, and long-term cardiovascular complications. Additionally, this review aims to identify potential therapeutic approaches to intervene with the RAS and prevent the development of AKI. Recent findings Recent studies have provided increasing evidence of RAS alteration during sepsis, with systemic and local RAS disturbance, which can contribute to SA-AKI. Angiotensin II was recently approved for catecholamine resistant vasodilatory shock and has been associated with improved outcomes in selected patients. Summary SA-AKI is a common condition that can involve disturbances in the RAS, particularly the canonical angiotensin-converting enzyme (ACE) angiotensin-II (Ang II)/angiotensin II receptor 1 (AT-1R) axis. Increased renin levels, a key enzyme in the RAS, have been shown to be associated with AKI and may also guide vasopressor therapy in shock. In patients with high renin levels, angiotensin II administration may reduce renin concentration, improve intra-renal hemodynamics, and enhance signaling through the angiotensin II receptor 1. Further studies are needed to explore the role of the RAS in SA-AKI and the potential for targeted therapies.
Article
Full-text available
Background: Dipeptidyl peptidase-3 (DPP3) is a metallopeptidase which cleaves bioactive peptides, notably angiotensin II, and is involved in inflammation regulation. DPP3 has been proposed to be a myocardial depressant factor and to be involved in circulatory failure in acute illnesses, possibly due to angiotensin II cleavage. In this study, we evaluated the association between plasmatic DPP3 level and outcome (mortality and hemodynamic failure) in severely ill burn patients. Methods: In this biomarker analysis of a prospective cohort study, we included severely ill adult burn patients in two tertiary burn intensive care units. DPP3 was measured at admission (DPP3admin) and 3 days after. The primary endpoint was 90-day mortality. Secondary endpoints were hemodynamic failure and acute kidney injury (AKI). Results: One hundred and eleven consecutive patients were enrolled. The median age was 48 (32.5-63) years, with a median total body surface area burned of 35% (25-53.5) and Abbreviated Burn Severity Index (ABSI) of 8 (7-11). Ninety-day mortality was 32%. The median DPP3admin was significantly higher in non-survivors versus survivors (53.3 ng/mL [IQR 28.8-103.5] versus 27.1 ng/mL [IQR 19.4-38.9]; p < 0.0001). Patients with a sustained elevated DPP3 had an increased risk of death compared to patients with high DPP3admin but decreased levels on day 3. Patients with circulatory failure had higher DPP3admin (39.2 ng/mL [IQR 25.9-76.1] versus 28.4 ng/mL [IQR 19.8-39.6]; p = 0.001) as well as patients with AKI (49.7 ng/mL [IQR 30.3-87.3] versus 27.6 ng/mL [IQR 19.4-41.4]; p = 0.001). DPP3admin added prognostic value on top of ABSI (added chi2 12.2, p = 0.0005), Sequential Organ Failure Assessment (SOFA) score at admission (added chi2 4.9, p = 0.0268), and plasma lactate at admission (added chi2 6.9, p = 0.0086) to predict circulatory failure within the first 48 h. Conclusions: Plasma DPP3 concentration at admission was associated with an increased risk of death, circulatory failure, and AKI in severely burned patients. Whether DPP3 plasma levels could identify patients who would respond to alternative hemodynamic support strategies, such as intravenous angiotensin II, should be explored.
Article
Full-text available
Aims: Acute heart failure is a high mortality disease and its pathophysiology is not completely understood. Dipeptidyl peptidase 3 (DPP3) is a cytosolic enzyme involved in angiotensin II and enkephalins cleavage. The aim of this study was to investigate the association of circulating DPP3 (cDPP3) levels and mortality in cardiogenic shock patients and to determine the effects of high cDPP3 on organ function in a heart failure (HF) model in mice. Methods and results: cDPP3 was measured in 174 patients in cardiogenic shock and high cDPP3 levels were associated with an increased short-term mortality risk (standardized hazard ratio: 1.4 (1.1-1.8)) and severe organ dysfunction. Additionally, a rapid decrease in cDPP3 in cardiogenic shock patients within 24 h of admission was associated with a favourable outcome. This study showed that injection of DPP3 induced myocardial depression (-10 ± 2% of shortening fraction) and impaired kidney haemodynamics (+0.30 ± 0.02 of renal resistive index) in healthy mice. cDPP3 inhibition by Procizumab, a specific antibody directed against cDPP3, promptly normalized cardiac function and kidney haemodynamics in an acute heart failure mouse model, with a marked reduction in oxidative stress and inflammatory signalling. Conclusion: Our study demonstrated cDPP3 is a newly discovered myocardial depressant factor, the levels of which at admission are associated with mortality in severe HF patients. Furthermore, inhibition of cDPP3 by Procizumab improved haemodynamics in a mouse model of HF. Our results suggest that DPP3 could be a new biomarker and biotarget for severe HF.
Article
Full-text available
Aims: Dipeptidyl peptidase 3 (DPP3) is a protease involved in the degradation of cardiovascular mediators. Its administration has been shown to be associated with impaired cardiac contraction and kidney haemodynamics while its inhibition restored cardiac contraction in a pre-clinical model of severe heart failure in mice. Circulating DPP3 (cDPP3) was found to be elevated in shock. The present study aims to assess the association between cDPP3 and worsening haemodynamics, namely refractory shock, in a cohort of cardiogenic shock (CS). Methods and results: This is an ancillary study of OptimaCC, a prospective, double-blind, multicentre, randomized study assessing efficacy and safety of catecholamines in 57 patients with CS after acute myocardial infarction. cDPP3 was measured in plasma at inclusion, 24 h, 48 h, and 72 h, and haemodynamic and biological parameters were recorded at inclusion. cDPP3 values were higher in refractory CS than non-refractory CS at inclusion (median [interquartile range]; 76.1 [37.9-238.7] ng/mL vs. 32.8 [23.9-47.6] ng/mL, P = 0.014), at 24 h (P < 0.001) and up to 48 h (P = 0.027). Furthermore, cDPP3 at inclusion discriminated CS patients who did develop refractory shock vs. non-refractory with an area under the curve of 0.73 (95% confidence interval [CI] 0.55-0.92). The high cDPP3 group (cDPP3 ≥59.1 ng/mL) at inclusion had a higher Simplified Acute Physiology Score II (SAPS II), lower cardiac index and lower estimated glomerular filtration rate. More importantly, in CS patients with high cDPP3 at inclusion, those who rapidly decreased cDPP3 at 24 h exhibited a striking reduction in the occurrence of refractory shock and death. Conclusion: In CS patients, cDPP3 gives an early prediction of outcome, including development of refractory status and/or survival. Clinical trial registration: clinicaltrials.gov Identifier NCT01367743.
Article
Full-text available
Background: The ubiquitously expressed dipeptidyl peptidase 3 (DPP3) is involved in protein metabolism, blood pressure regulation, and pain modulation. These diverse functions of DPP3 are attributed to the degradation of bioactive peptides like angiotensin II. However, because of limitations in currently available assays for determination of active DPP3 in plasma, the exact physiological function of DPP3 and its role in the catabolism of bioactive peptides is understudied. Here, we developed 2 assays to specifically detect and quantify DPP3 protein and activity in plasma and validated DPP3 quantification in samples from critically ill patients. Methods: Assay performance was evaluated in a sandwich-type luminometric immunoassay (LIA) and an enzyme capture activity assay (ECA). DPP3 plasma concentrations and activities were detected in a healthy, population-based cohort and in critically ill patients suffering from severe sepsis and septic shock. Results: The DPP3-LIA and DPP3-ECA show an almost ideal correlation and very similar and robust performance characteristics. DPP3 activity is detectable in plasma of predominantly healthy subjects with a mean (±SD) of 58.6 (±20.5) U/L. Septic patients show significantly increased DPP3 plasma activity at hospital admission. DPP3 activities further increase in patients with more severe conditions and high mortality risk. Conclusion: We developed 2 highly specific assays for the detection of DPP3 in plasma. These assays allow the use of DPP3 as a biomarker for the severity of acute clinical conditions and will be of great value for future investigations of DPP3's role in bioactive peptide degradation in general and the angiotensin II pathway in specific.
Article
Full-text available
Bacteroides thetaiotaomicron is a dominant member of the human intestinal microbiome. The genome of this anaerobe encodes more than 100 proteolytic enzymes, the majority of which have not been characterized. In the present study, we have produced and purified recombinant dipeptidyl peptidase III (DPP III) from B. thetaiotaomicron for the purposes of biochemical and structural investigations. DPP III is a cytosolic zinc-metallopeptidase of the M49 family, involved in protein metabolism. The biochemical results for B. thetaiotaomicron DPP III from our research showed both some similarities to, as well as certain differences from, previously characterised yeast and human DPP III. The 3D-structure of B. thetaiotaomicron DPP III was determined by X-ray crystallography and revealed a two-domain protein. The ligand-free structure (refined to 2.4 Å) was in the open conformation, while in the presence of the hydroxamate inhibitor Tyr-Phe-NHOH, the closed form (refined to 3.3 Å) was observed. Compared to the closed form, the two domains of the open form are rotated away from each other by about 28 degrees. A comparison of the crystal structure of B. thetaiotaomicron DPP III with that of the human and yeast enzymes revealed a similar overall fold. However, a significant difference with functional implications was discovered in the upper domain, farther away from the catalytic centre. In addition, our data indicate that large protein flexibility might be conserved in the M49 family.
Article
Full-text available
Background Reliable measurement of blood pressure in conscious mice is essential in cardiovascular research. Telemetry, the “gold‐standard” technique, is invasive and expensive and therefore tail‐cuff, a noninvasive alternative, is widely used. However, tail‐cuff requires handling and restraint during measurement, which may cause stress affecting blood pressure and undermining reliability of the results. Methods and Results C57Bl/6J mice were implanted with radio‐telemetry probes to investigate the effects of the steps of the tail‐cuff technique on central blood pressure, heart rate, and temperature. This included comparison of handling techniques, operator's sex, habituation, and influence of hypertension induced by angiotensin II. Direct comparison of measurements obtained by telemetry and tail‐cuff were made in the same mouse. The results revealed significant increases in central blood pressure, heart rate, and core body temperature from baseline following handling interventions without significant difference among the different handling technique, habituation, or sex of the investigator. Restraint induced the largest and sustained increase in cardiovascular parameters and temperature. The tail‐cuff readings significantly underestimated those from simultaneous telemetry recordings; however, “nonsimultaneous” telemetry, obtained in undisturbed mice, were similar to tail‐cuff readings obtained in undisturbed mice on the same day. Conclusions This study reveals that the tail‐cuff technique underestimates the core blood pressure changes that occur simultaneously during the restraint and measurement phases. However, the measurements between the 2 techniques are similar when tail‐cuff readings are compared with telemetry readings in the nondisturbed mice. The differences between the simultaneous recordings by the 2 techniques should be recognized by researchers.
Article
Full-text available
NRF2 is a transcription factor serving as a master regulator of the expression of many genes involved in cellular responses to oxidative and other stresses. In the absence of stress, NRF2 is constantly synthesized but maintained at low levels as it is targeted by KEAP1 for ubiquitination and proteasome-mediated degradation. NRF2 binds KEAP1 mainly through a conserved "ETGE" motif that has also been found in several other proteins, such as DPP3, which has been shown to bind KEAP1 and enhance NRF2 function upon overexpression. Here we demonstrate the interaction between endogenous DPP3 and endogenous KEAP1. We further show that the DPP3-KEAP1 interaction is strongly induced by hydrogen peroxide and that DPP3 is required for timely NRF2 induction and nuclear accumulation in the estrogen receptor (ER)-positive MCF7 breast cancer cells. Moreover, we present evidence that the binding of DPP3 to KEAP1 stabilizes the latter. Finally, we show that DPP3 is overexpressed in breast cancer and that elevated levels of DPP3 mRNA correlate with increased NRF2 downstream gene expression and poor prognosis, particularly for ER-positive breast cancer. Our studies reveal novel insights into the regulation of NRF2 and identify DPP3 and an NRF2 transcriptional signature as potential biomarkers for breast cancer prognosis and treatment.
Article
Controlling oxidative stress through the activation of antioxidant pathways is crucial in bone homeostasis and impairments of the cellular defense systems involved contribute to the pathogenesis of common skeletal diseases. Here we focused on the dipeptidyl peptidase 3 (DPP3), a poorly investigated ubiquitous zinc‐dependent exopeptidase activating the Keap1‐Nrf2 antioxidant pathway. We demonstrated Dpp3 expression in bone and, to understand its role in this compartment, we generated a Dpp3 knockout (KO) mouse model and specifically investigated the skeletal phenotype. Adult Dpp3 KO mice showed a mild growth defect, a significant increase in bone marrow cellularity and bone loss mainly caused by increased osteoclast activity. Overall, in the mouse model lack of DPP3 resulted in sustained oxidative stress and in alterations of bone microenvironment favoring the osteoclast compared to the osteoblast lineage. Accordingly, in vitro studies revealed that Dpp3 KO osteoclasts had an inherent increased resorptive activity and ROS production, which on the other hand made them prone to apoptosis. Moreover, absence of DPP3 augmented bone loss after estrogen withdrawal in female mice, further supporting its relevance in the framework of bone pathophysiology. Overall, we demonstrated a non‐redundant role for DPP3 in the maintenance of bone homeostasis and proposed that DPP3 might represent a possible new osteoimmunological player and a marker of human bone loss pathology. This article is protected by copyright. All rights reserved.
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 blockades of this system by the inhibition of angiotensin-converting enzyme (ACE) or antagonism of the angiotensin type 1 receptor (AT1R) are effective therapeutic regimens. The RAS is now defined as a 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 nonclassical or alternative RAS is composed primarily of the ACE2-Ang-(1-7)-AT7R pathway that opposes the Ang II-AT1R axis. In lieu of the complex aspects of this system, the current review assesses the enzymatic cascade of the alternative Ang-(1-7) axis of the RAS.
Article
The overproduction of reactive oxygen species (ROS) generates oxidative stress in cells. Oxidative stress results in various pathophysiological conditions, especially cancers and neurodegenerative diseases (NDD). The Keap1–Nrf2 [Kelch-like ECH-associated protein 1–nuclear factor (erythroid-derived 2)-like 2] regulatory pathway plays a central role in protecting cells against oxidative and xenobiotic stresses. The Nrf2 transcription factor activates the transcription of several cytoprotective genes that have been implicated in protection from cancer and NDD. The Keap1–Nrf2 system acts as a double-edged sword: Nrf2 activity protects cells and makes the cell resistant to oxidative and electrophilic stresses, whereas elevated Nrf2 activity helps in cancer cell survival and proliferation. Several groups in the recent past, from both academics and industry, have reported the potential role of Nrf2-mediated transcription to protect from cancer and NDD, resulting from mechanisms involving xenobiotic and oxidative stress. It suggests that the Keap1–Nrf2 system is a potential therapeutic target to combat cancer and NDD by designing and developing modulators (inhibitors/activators) for Nrf2 activation. Herein, we review and discuss the recent advancement in the regulation of the Keap1–Nrf2 system, its role under physiological and pathophysiological conditions including cancer and NDD, and modulators design strategies for Nrf2 activation.