Jpet 122 001339 Full

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 41

JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.

001339
This article has not been copyedited and formatted. The final version may differ from this version.

Is diminazene an angiotensin-converting enzyme 2 (ACE2) activator? Experimental


evidence and implications

Shiva Hadi Esfahani1, Srinidhi Jayaraman1, Vardan T. Karamyan1,2


1
Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health
Sciences Center, Amarillo, USA
2
Current affiliation: Department of Foundational Medical Studies, Oakland University William
Beaumont School of Medicine, Rochester, MI, USA

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022

1
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

Running title: Diminazene is not an ACE2 activator.

Corresponding author:
Vardan T. Karamyan, Pharm.D., Ph.D.
1300 Coulter St., Amarillo, TX 79106
Email: vardan.karamyan@ttuhsc.edu
Phone: 806-414-9239

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


Number of manuscript pages: 33

Number of tables: 1

Number of figures: 7

Number of references: 50

Number of words in the Abstract: 237

Number of words in the Introduction: 448

Number of words in the Discussion: 1602

Abbreviations: angiotensin-converting enzyme 2 (ACE2), angiotensin-(1-7) (Ang-(1-7)),


angiotensin II (Ang II), diminazene aceturate (DIZE), neprilysin (NEP), neurolysin (Nln), thimet
oligopeptidase (TOP), 2-(N-Morpholino)ethanesulfonic acid (MES), liquid chromatography
mass spectrometry (LCMS), artificial cerebrospinal fluid (aCSF), fluorescence unit (FLU), a
disintegrin and metalloproteinase 17 (ADAM17), SARS‐coronavirus (SARS‐CoV).

Recommended section assignment: Drug Discovery and Translational Medicine

2
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

Abstract

Antiprotozoal veterinary drug diminazene aceturate (DIZE) has been proposed to be an

angiotensin-converting enzyme 2 (ACE2) activator. Since then, DIZE was used in dozens of

experimental studies, but its mechanism of action attributed to ACE2 activation and enhanced

formation of angiontensin-(1-7) (Ang-(1-7)) from Ang II was not carefully verified. The aim of

this study was to confirm the effect of DIZE on catalytic activity of ACE2 and extend it to other

peptidases involved in formation and degradation of Ang-(1-7). Concentration-dependent effect

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


of DIZE on the initial rate of a fluorogenic substrate hydrolysis by human and mouse

recombinant ACE2 was measured at assay conditions imitating that of the original report but no

activation of ACE2 was documented. Similar results were obtained with a more physiologically

relevant assay buffer. In addition, DIZE did not affect activity of recombinant neprilysin,

neurolysin, thimet oligopeptidase and ACE. Efficiency of the fluorogenic substrate hydrolysis

(Vmax/Km value) by ACE2 in response to different concentrations of DIZE was also measured but

no substantial effects were documented. Likewise, DIZE failed to enhance the hydrolysis of

ACE2 endogenous substrate Ang II. Identity of the commercial recombinant ACE2 variants used

in these experiments was confirmed by inhibition with two well-characterized inhibitors (DX600

and MLN4760), activation by NaCl and Western Blotting using validated antibodies. These

observations challenge the widely accepted notion about the molecular mechanism of DIZE

action and call for not ascribing this molecule as an ACE2 activator.

3
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

Significance statement

Diminazene aceturate (DIZE) has been proposed and widely used in experimental studies as an

angiotensin-converting enzyme 2 (ACE2) activator. Our detailed in vitro pharmacological

studies failed to confirm that DIZE is an ACE2 activator. In addition, DIZE did not substantially

affect the activity of other peptidases involved in formation and degradation of angiotensin-(1-7).

Researchers should refrain from calling DIZE an ACE2 activator. Other mechanisms are

responsible for the therapeutic benefits attributed to DIZE.

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022

4
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

Introduction

Angiotensin-converting enzyme 2 (ACE2) is a critical member of the renin-angiotensin system

(RAS), responsible for activation of angiotensin-(1-7)/Mas receptor axis and counteracting many

of the pathophysiological consequences of ACE/angiotensin II/angiotensin type 1 receptor axis

(Bader et al., 2020). Since its discovery by two independent groups in 2000 (Donoghue et al.,

2000; Tipnis et al., 2000), key functions of ACE2 and its critical role in pathogenesis of various

cardio- and cerebrovascular, renal and endocrine disorders have been studied by multiple

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


research groups (Marquez et al., 2021). Focus on ACE2 was further enhanced during mid-2000

by a discovery that the peptidase serves as a receptor for SARS‐coronavirus (SARS‐CoV) (Li et

al., 2003) and most recently, by the ongoing COVID-19 pandemic caused by SARS‐CoV-2 virus

which also uses ACE2 to enter host cells (Hoffmann et al., 2020). This increased focus on ACE2

has led to many exciting discoveries related to understanding of various disorders and pathogenic

mechanisms, as well as molecular intricacies of catalysis and modulation of ACE2 and related

peptidases. Given the importance of ACE2 in the function of RAS, a number of approaches have

been explored to harness the therapeutic potential of this enzyme, including use of recombinant

ACE2 as a biologic agent (Marquez et al., 2021). Among these studies, an intriguing and

potentially clinically impactful discovery was identification of a small molecule activator of

ACE2, diminazene (DIZE) (Kulemina and Ostrov, 2011). Diminazene aceturate is an

antiprotozoal agent primarily used in cattle and small ruminants but it is not approved for use in

humans by the FDA (Kuriakose and Uzonna, 2014). Since the original report suggesting

stimulation of ACE2 activity by DIZE, few dozen research groups used this pharmacological

agent for proof-of-concept studies to document beneficial function of ACE2 in preventing and/or

reversing various disease conditions, including myocardial infarction (Qi et al., 2013; Castardeli

5
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

et al., 2018), pulmonary hypertension (Rigatto et al., 2013; Shenoy et al., 2013), ischemic stroke

(Bennion et al., 2015) and Alzheimer’s disease (Kamel et al., 2018; Evans et al., 2020). Despite

the widespread use of DIZE as an ACE2 activator in experimental studies, little was known

about selectivity of this agent towards peptidases that belong to the same family of enzymes as

ACE2 and are involved in formation and degradation of angiotensin-(1-7) (Ang-(1-7)).

Therefore, the original objective of this study was to confirm enhanced catalytic activity of

ACE2 in response to DIZE and evaluate the effect of this drug on activity of neprilysin (NEP),

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


neurolysin (Nln), thimet oligopeptidase (TOP) and ACE. Here, we report the results of detailed

in vitro pharmacological studies which indicate that DIZE is not a direct activator of ACE2 and

challenge the previous observations and widely accepted notion about this molecule.

6
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

Materials and Methods

Materials – DIZE was purchased from Sigma Aldrich Co. (product D7770-1G, lots SLBS6137

and SLBZ7521). Recombinant human and mouse ACE2 (products 933-ZN and 3437-ZN),

human NEP (product 1182-ZNC), human Nln (product 3814-ZN), human TOP (product 3439-

ZN) and human ACE (product 929-ZN) were obtained from R&D Systems (Minneapolis, MN).

Selective ACE2 inhibitors, DX600 (product AS-62337) and MLN4760 (product 5306160001)

were purchased from AnaSpec and Millipore-Sigma, respectively. Fluorogenic substrate Mca-

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


Pro-Leu-Gly-Pro-D-Lys(Dnp)-OH (product 4027687) was obtained from Bachem, whereas Mca-

Arg-Pro-Pro-Gly-Phe-Ser-Ala-Phe-Lys(Dnp) and Mca-Ala-Pro-Lys(Dnp) were purchased from

Enzo Life Sciences (products BML-P227 and BML-P163).

Enzymatic assays – Activity of peptidases was measured in a continuous assay by documenting

the increase in fluorescence signal in the result of hydrolysis of a quenched fluorescence

substrate as detailed in our previous publications (Wangler et al., 2016; Jayaraman et al., 2020).

For ACE2 we used fluorogenic substrate Mca-Ala-Pro-Lys (Dnp) (Kulemina and Ostrov, 2011),

for ACE and NEP, Mca-Arg-Pro-Pro-Gly-Phe-Ser-Ala-Phe-Lys(Dnp)-OH (Johnson and Ahn,

2000; Wösten-van Asperen et al., 2008) and for Nln and TOP, Mca-Pro-Leu-Gly-Pro-D-

Lys(Dnp)-OH (Jayaraman et al., 2021). As detailed in results section, the first set of experiments

were carried out in 75 mM Tris-HCl 0.1 M NaCl, 0.5 µM ZnSO4 and 0.01% Triton-X (pH 7.4)

which is the same assay buffer used in the original study reporting discovery of DIZE as an

ACE2 activator (Kulemina and Ostrov, 2011). To mimic a more physiologically relevant

environment, specifically that of the brain due to our long-standing interests in brain peptidases

(Karamyan and Speth, 2007; Karamyan et al., 2009; Rashid et al., 2014; Wangler et al., 2016),

7
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

subsequent experiments were carried out in artificial cerebrospinal fluid (aCSF; 0.1% Triton X‐

100, 126 mM NaCl, 26 mM NaHCO3 , 3 mM KCl, 4 mM glucose, 1.4 mM KH2PO4 , 25 mM

HEPES, 1.4 mM CaCl2 , 1.3 mM MgCl2, 0.2 M ZnSO4 , pH 7.2). In experiments with TOP, the

assay buffer additionally contained 0.1 mM dithiothreitol (Shrimpton et al., 1997). DIZE was

dissolved in deionized water to make 30 mM stock followed by dilution in the assay buffer for

working stocks. DIZE (0.1 to 100 M in 100 µl assay volume in 96-well plates) was incubated

with recombinant ACE2 (0.05 or 0.065 nM human, and 0.04 or 0.065 nM mouse ACE2), ACE

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


(0.2 or 0.5 nM), Nln (0.3 or 1.2 nM), NEP (0.12 or 0.8 nM) or TOP (0.3 or 1.2 nM) for 10

minutes at 37C before addition of the respective fluorogenic substrate to start the reaction. In all

enzymatic assays, generation of the fluorescent product was documented every 1 min in a plate

reader (ex = 320, em = 405; SynergyMX; Biotek) at initial velocity conditions where ~10% of

the substrate was hydrolyzed and each sample was present in duplicate.

The inherent fluorescence quenching or enhancing property of DIZE was tested under the same

experimental conditions, with the exception that 2 µM assay concentration of Mca-Pro-Leu-OH

(hydrolysis product of the quenched fluorescence substrate; ex = 320, em = 405), was present

with ACE2 and different concentrations of DIZE.

NaCl dependency of ACE2 activity was measured in the same way as detailed above, except that

the assay buffer was 50 mM MES, 10 µM ZnSO4, 0.01% Triton X-100, pH 6.5 with additional 0,

0.1, 0.5 or 1 M NaCl (Vickers et al., 2002).

8
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

Kinetic experiments and enzyme inhibition – Kinetic parameters for both recombinant human and

mouse ACE2 in the presence of 10 and 50 μM assay concentrations of DIZE were determined by

addition of 2.5 to 100 µM fluorogenic substrate. DIZE was preincubated with human ACE2

(0.12 nM) or mouse (0.06 or 0.12 nM) ACE2 in aCSF at 37°C for 10 min before the addition of

the fluorogenic substrate as described above. In inhibition assays involving DX600 and

MLN4760, increasing concentrations (0.1 nM – 1 µM) of either inhibitor was preincubated with

human (0.08 nM) or mouse (0.065 or 0.09 nM) ACE2 in aCSF at 37°C for 10 min followed by

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


addition of the fluorogenic substrate.

Measurement of Ang-(1-7) formation from Ang II by liquid chromatography-mass spectrometry

analysis – Recombinant human or mouse ACE2 (0.3 nM and 0.5 nM, respectively) were

incubated with 10 or 50 μM DIZE in aCSF at 37°C for 10 min (30 µl assay volume), followed by

addition of 20 μM angiotensin II (Ang II, product 002-12, Phoenix Pharmaceuticals, Inc.) and

further incubation at 37°C for 10 min. These conditions ensured no more than 15% hydrolysis of

Ang II. The reaction was stopped by addition of 1 µl of 1.0 N LCMS grade HCl and storage at

−80°C freezer. Quantification of Ang II and its hydrolysis product Ang-(1-7) was carried out by

LC-MS/MS using a SCIEX Triple Quad™ 5500+ LC-MS/MS System following a protocol

modified from our recent studies (Jayaraman et al., 2021; Rahman et al., 2021). In brief, 2 μl of

sample was injected onto a 50 × 2.1 mm Kinetex EVO C18 column (Phenomenex). DAMGO, a

synthetic opioid (product 1171, Tocris Bioscience), was used as an internal standard and was

spiked into each sample. Chromatography was performed at flow rate of 300 μl min-1 and total

run time of 10 min, mobile phases consisted of 0.1% formic acid in water (solvent A) and 0.1%

formic acid in acetonitrile (solvent B). The gradient of solvent B was: linear increase from 3 to

9
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

35% during the first 5 min, increase to 85% in the next 50 sec, continuation at this level until 7

min, return to 3% in the next 50 sec and continuation at this level until 10 min. Detection of Ang

II and Ang-(1-7) were carried out in the positive polarity and multiple‐reaction monitoring mode

(150 msec dwell times and 5500 volts). The collision energy used for Ang II and Ang-(1-7),

were 29 and 30 volts, respectively. The precursor ion of charged state was calculated by mass-to-

charge ratio (m/z), as M + 1, (M + 2)/2 or (M + 3)/3 primary ions, and identified and fragmented

by a collision-induced dissociation (CID) voltage. The Q1 to Q3 transitions were m/z 524.1

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


precursor ion to m/z 263.1 for Ang II, m/z 450.4 precursor ion to m/z 110.1 for Ang-(1-7), and

m/z 514 precursor ion to m/z 134.1 for DAMGO. The peak areas for Ang II and Ang-(1-7) and

the internal standard were obtained through Analyst software™ (version 1.7.2) and expressed as

peak area ratio (peptide peak area/internal standard peak area).

Western Blotting – To confirm the identity of the recombinant human and mouse ACE2

purchased from a commercial vendor, a set of Western Blotting experiments were carried out.

For this, human (20 and 40 ng) and mouse (40 and 80 ng) ACE2, and human ACE (negative

control, 20 and 40 ng) were resolved in a 7.5% precast polyacrylamide gel (Mini-PROTEAN

TGX, product 4561023, Bio-Rad Laboratories) using conventional SDS/PAGE as described in

our previous publications (Al Shoyaib et al., 2021a; Al Shoyaib et al., 2021b). The resolved

samples were transferred onto a polyvinylidene difluoride (PVDF) membrane (product 88518,

Thermo Scientific), blocked with 1% BSA (product BP9703-199, Fisher Scientific) solution in

Tris-buffered saline with Tween-20 (TBST) and incubated with rabbit polyclonal anti-ACE2

antibody (freshly diluted in TBST containing 1% BSA at 1:10000; product RP2-ACE-2, Triple

Point Biologics Inc.; also available from Abcam, product AB-38941) at room temperature

10
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

overnight. Next, the membrane was washed five times with TBST and incubated with

horseradish peroxidase conjugated goat anti-rabbit secondary antibody (freshly diluted in TBST

containing 5% non-fat dry milk at 1:10000 dilution; product 170-6515, Bio-Rad laboratories) for

45 min. Afterwards, the membrane was washed with TBST, incubated with Super SignalTM West

Pico PLUS chemiluminescent detection reagent (Thermo Scientific) and exposed to an X-ray

film. Next, the membrane was incubated in Restore PLUS Western Blot Stripping Buffer

(product 46430, Thermo Scientific) for 15 minutes at room temperature, washed and blocked as

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


noted above, and incubated with rabbit polyclonal anti-ACE antibody (freshly diluted in TBST

containing 1% BSA at 1:20000; product RP3-ACE, Triple Point Biologics Inc.; also available

from Abcam, product AB-39172) at room temperature overnight. All subsequent steps to detect

ACE via chemiluminescence were carried out as detailed above, with an exception that the

secondary antibody was used at 1:20000 dilution. The original images of the films were cropped

and labeled using Photoshop.

Statistical analyses – Statistical analyses and curve fitting were carried out using GraphPad

Prism 9.3.1 software. The liner regression model of the software (V0 = Slope =  fluorescent

intensity of the reaction product/ time) was used to calculate slope of the line for each

enzymatic reaction, which represents the initial velocity (V0) for the reaction progress curve. For

calculation of A50 and Amax values, V0 values for hydrolysis of the fluorogenic substrate in the

presence of varying concentrations of DIZE were expressed as percent within each experiment,

100% being V0 of enzymatic reaction in the absence of DIZE (i.e., basal activity with vehicle

control). Curve fitting of %V0 was done by a nonlinear regression model for the three-parameter

log(stimulator) vs. response equation [Y=Bottom + (Top-Bottom)/(1+10^((LogA50-X))]. Km

11
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

and Vmax values were calculated by fitting initial velocity values for hydrolysis of varying

concentrations of the fluorogenic substrate by ACE2 in the absence or presence of 10 or 50 µM

of DIZE into Michaelis-Menten equation [Y=Vmax*X/(Km + X)]. IC50 values for selective

ACE2 inhibitors were calculated by fitting initial velocity values for hydrolysis of the

fluorogenic substrate by ACE2 in the presence of varying concentrations of each inhibitor into a

nonlinear regression model for the three-parameter log(inhibitor) vs. response equation

[Y=Bottom + (Top-Bottom)/(1+10^((X-LogIC50))]. Ki values were calculated based on the

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


Cheng-Prusoff equation: Ki=IC50/(1+S/Km) where S is the substrate concentration (10 µM for

the fluorogenic substrate in our experiments), and Km is the Km value for the substrate (19.4 and

9.8 µM for human and mouse ACE2 respectively, see Table 1). Comparison of data from

experiments with several groups (Figs. 3, 4 and 6) was done by one-way ANOVA followed by

Dunnett’s multiple comparison test. A p-value < 0.05 was considered statistically significant.

Data are presented as mean, mean with 95% confidence intervals or mean ± S.D.

12
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

Results

Effect of DIZE on activity of human and mouse recombinant ACE2 in Tris buffer – To replicate

activation of human recombinant ACE2 by DIZE, we used the same experimental conditions

(including Tris HCl assay buffer, fluorogenic substrate, ACE2, etc.) reported by Kulemina and

Ostrov (2011). Concentration-dependent effect of DIZE on initial velocity of fluorogenic

substrate hydrolysis by ACE2 is presented in Fig. 1, indicating negligible changes in activity of

the peptidase with up to 30 µM DIZE, and 20.5% (95% CI, 16.4 – 24.5%) inhibition with 100

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


µM of the drug. Since progression of the enzymatic reaction in these experiments was evaluated

by tracking the generated fluorescence signal, we additionally evaluated the effect of DIZE on

fluorescence properties of Mca (7-methoxycoumarin-4-acetyl) group in Mca-Pro-Leu-OH under

the same experimental conditions. It was revealed that at high concentrations DIZE has

fluorescence quenching properties, which amounts to 18.6% (95% CI, 17.9 – 19.3%) at 100 µM

(Fig. 1) and is more pronounced at higher concentrations of the drug.

We repeated the same experiments with mouse recombinant ACE2 and observed nearly identical

results – no changes in activity of the peptidase with up to 30 µM DIZE, 28.9% (95% CI, 20.2 –

37.3%) inhibition with 100 µM of the drug, and 17.6% (95% CI, 16.6 – 18.5%) quenching of the

fluorescence signal in the presence of 100 µM DIZE (Fig. 1).

Notably, the lack of DIZE effect on ACE2 activity was contrary to our expectation because of

which, we evaluated two different lots of DIZE and ACE2 but documented similar results (the

complied data are presented in this manuscript).

Effects of DIZE on activity of human and mouse recombinant ACE2 in aCSF – In this set of

experiments, we repeated the approach described above using aCSF instead of Tris buffer for

13
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

assays, because of our long-standing interests in brain peptidases and a need to study ACE2 in a

more physiologically relevant environment. In this case too, DIZE inhibited activity of human

and mouse ACE2 by 13.8% (95% CI, 12.2 – 15.3%) and 30.3% (95% CI, 27.2 – 33.3%) at 100

µM, and had negligible effect at lower concentrations (Fig. 2).

Effects of DIZE on activity of other peptidases involved in formation and degradation of Ang-(1-

7) – To understand whether DIZE may affect activity of other peptidases related to ACE2 and by

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


that modulate availability of Ang-(1-7), we studied its effects on catalytic activity of human

recombinant NEP (neprilysin, aka neutral endopeptidase), Nln (neurolysin), TOP (thimet

oligopeptidase) and ACE (Fig. 2). DIZE had negligible effect on activity of these peptidases at

low concentrations, but at 100 µM it inhibited activity of NEP by 21.4% (95% CI, 17.8 –

25.7%), Nln by 18.7% (95% CI, 15.9 – 21.4%), TOP by 18.6% (95% CI, 13.1 – 24.2%), and

ACE by 27.6% (95% CI, 23.9 – 31.4%).

Effect of DIZE on catalytic efficiency of ACE2 – To determine whether DIZE may increase the

catalytic efficiency of ACE2 without apparent increase in initial velocity of the substrate

hydrolysis, in this set of experiments we studied its effect on hydrolysis of synthetic substrate at

different concentrations. Since half-maximal activating concentration (A50) of DIZE reported by

Kulemina and Ostrov (2011) was 8 µM with Amax value of ~300%, we used 10 and 50 µM assay

concentrations of DIZE to observe unambiguous effects. In our experiments, the maximal

velocity (Vmax) for human ACE2 was increased by ~2% in the presence of 10 µM DIZE and

decreased by ~25% in the presence of 50 µM DIZE (Fig. 3 and Table 1), with respective ~11%

increase and ~35% decrease in calculated Km values. Similarly, with mouse ACE2 the calculated

14
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

Vmax values decreased by ~10% and 25% in the presence of 10 and 50 µM DIZE, whereas Km

values decreased by ~2 and 14% (Fig. 3 and Table 1).

As summarized in Table 1, we observed an overall decrease in catalytic efficiency (Vmax/Km

value) of ACE2 in the presence of DIZE, with exception of the human variant in response to 50

µM of DIZE where ~17% increase was documented.

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


Effect of DIZE on ACE2-mediated formation of Ang-(1-7) from Ang II – Since observations made

with synthetic substrates may be misleading in identification of enzyme modulators (Jayaraman

et al., 2021), in this set of experiments the concentration-dependent effect of DIZE on hydrolysis

of Ang II by ACE2 was studied using mass spectrometry and no substantial differences were

documented (Fig. 4). More specifically, in case of human ACE2 the relative amount (peak area

of Ang-(1-7)/internal standard) of generated Ang-(1-7) in the absence of DIZE (i.e., vehicle

control) was 0.32 (95% CI, 0.29 – 0.33), whereas it was 0.30 (95% CI, 0.27 – 0.32) and 0.30

(95% CI, 0.28 – 0.32) in the presence of 10 and 50 µM DIZE, respectively. Similarly, in case of

mouse ACE2 the relative amount of formed Ang-(1-7) in the absence of DIZE was 0.61 (95%

CI, 0.59 – 0.63), whereas it was 0.60 (95% CI, 0.58 – 0.62) and 0.59 (95% CI, 0.57 – 0.61) in the

presence of 10 and 50 µM DIZE, respectively (Fig. 4). In line with this observation, the relative

amount of Ang II (peak area of Ang II/internal standard) in the same samples for human ACE2

was 1.68 (95% CI, 1.64 – 1.72; vehicle), 1.68 (95% CI, 1.65 – 1.71; 10 µM DIZE) and 1.69

(95% CI, 1.65 – 1.73; 50 µM DIZE), whereas for mouse ACE2 it was 1.53 (95% CI, 1.49 – 1.56;

vehicle), 1.54 (95% CI, 1.5 – 1.58; 10 µM DIZE) and 1.53 (95% CI, 1.49 – 1.57; 50 µM DIZE)

(Fig. 4). As expected, the Ang-(1-7)/Ang II ratio (i.e. ratio of relative amounts) was also similar

15
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

in these samples, being 0.187 (95% CI, 0.177 – 0.198; vehicle), 0.176 (95% CI, 0.162 – 0.189;

10 µM DIZE) and 0.177 (95% CI, 0.162 – 0.192; 50 µM DIZE) for human ACE2, and 0.40

(95% CI, 0.391 – 0.410; vehicle), 0.387 (95% CI, 0.374 – 0.401; 10 µM DIZE) and 0.387 (95%

CI, 0.374 – 0.399; 50 µM DIZE) for mouse ACE2.

Effects of MLN4760 and DX600 on activity of human and mouse recombinant ACE2 – To verify

identities of human and mouse recombinant ACE2 used in the above-described experiments,

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


here we used two well-characterized selective inhibitors of the peptidase, MLN4760 and DX600,

to study the inhibition profile of the commercial ACE2. Concentration-dependent effects of both

inhibitors on initial velocity of fluorogenic substrate hydrolysis by human and mouse ACE2 are

presented in Fig. 5. In case of MLN4760, the calculated IC50 values are 0.51 nM (95% CI, 0.40 –

0.64 nM; Ki = 0.34 nM) and 8.5 nM (95% CI, 8.02 – 9.19 nM; Ki = 4.25 nM) for human and

mouse ACE2, respectively. In case of DX600, the calculated IC50 value is 41.7 nM (95% CI,

35.3 – 49.2 nM; Ki = 27.5 nM) for human ACE2, whereas for the mouse variant, as expected, we

observed low-potency inhibition (IC50 > 1 µM).

Effect of sodium chloride on activity of human and mouse recombinant ACE2 – One of the

interesting features of human ACE2 is the ability of NaCl to enhance its catalytic activity

(Vickers et al., 2002). To evaluate this feature of the commercial human ACE2 used in the

above-described experiments, we conducted a new set of assays using a MES-based buffer as

originally described. Similar to the earlier reports (Vickers et al., 2002; Guy et al., 2003), we

documented profoundly enhanced activity of human ACE2 in the presence of increasing

concentrations of NaCl. The initial velocity of hydrolysis (V0) was 122 (95% CI, 104 – 140) in

16
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

the absence of NaCl, and 329 (95% CI, 271 – 387), 616 (95% CI, 554 – 678), and 559 (95% CI,

510 – 609) in the presence of 0.1, 0.5 and 1 M NaCl. The same experiment was also conducted

with mouse ACE2, however only slight activation was observed with 0.1 M NaCl, and inhibition

was documented with 1 M NaCl; V0 values were 466 (95% CI, 430 – 501), 549 (95% CI, 501 –

598), 414 (95% CI, 374 – 454), and 326 (95% CI, 300 – 352) for 0, 0.1, 0.5 and 1 M NaCl (Fig.

6).

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


Confirmation of ACE2 identity by Western Blotting – In this last set of experiments, we used

commercial antibodies selective for ACE2 to verify the identity of the enzymes used in our study

by conventional Western Blotting. As expected, these experiments confirmed identity of the

recombinant ACE2 and specificity of the used anti-ACE2 antibody (Fig. 7).

17
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

Discussion

In this study, we sought to verify the direct effect of DIZE on activity of ACE2 and extended it

to closely related peptidases that are involved in formation and degradation of Ang-(1-7). DIZE,

which is an antiprotozoal agent used in veterinary medicine, was identified by Kulemina and

Ostrov (2011) to directly interact with ACE2 and enhance its catalytic activity. We replicated the

assay conditions of the original report, but contrary to our expectation, no activation of human

recombinant ACE2 was observed over a wide range of DIZE concentrations (Fig. 1). Since many

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


studies using DIZE as an ACE2 activator have been conducted in mice, we additionally did the

same experiment using mouse recombinant ACE2 but failed to document activation of the

peptidase. Similar observations were documented in aCSF which was used as a more

physiologically relevant assay buffer (Rashid et al., 2014) (Fig. 2). We also revealed, that at 100

µM and higher concentrations, DIZE quenches the fluorescence signal of the product generated

from the hydrolysis of fluorogenic substrate in these assays. This revelation indicates

overestimation of ACE2 inhibition observed with 100 µM DIZE (Figs. 1 and 2). Similarly,

negligible effects of DIZE were documented on activities of recombinant NEP, Nln, TOP and

ACE, peptidases involved in processing of angiotensins and Ang-(1-7) (Karamyan and Speth,

2007) (Fig. 2). We sought to clarify the concentration-dependent effect of DIZE on efficiency of

fluorogenic substrate hydrolysis by ACE2 (Vmax/Km value) but documented minor effects (Fig. 3

and Table 1) that are substantially different from the earlier reported observations (Kulemina and

Ostrov, 2011). In an effort to reproduce the original findings, we also conducted a set of

experiments using Ang II as a substrate. Regretfully, in this case too we did not observe

increased production of Ang-(1-7) by human or mouse ACE2 in the presence of DIZE (Fig. 4).

To exclude the possibility that we experimented on a different enzyme, we additionally

18
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

authenticated the recombinant ACE2 purchased from a commercial vendor. First, we used two

well-characterized selective ACE2 inhibitors and documented concentration-dependent

inhibition of human and mouse ACE2 with Ki values in line with previous reports (Fig. 5). These

experiments also confirmed an earlier finding that DX600 is a poor mouse ACE2 inhibitor

(Pedersen et al., 2011). Since NaCl is known to enhance activity of human ACE2 (Vickers et al.,

2002; Guy et al., 2003), next we evaluated this property of the peptidase. Similar to earlier

reports, we observed a profound increase in activity of human ACE2 in the presence of

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


increasing concentrations of NaCl, whereas there was a small activation or inhibition with mouse

ACE2 (Fig. 6). Currently, we do not know why NaCl does not enhance activity of mouse ACE2,

and we are not aware of another study investigating this question. Lastly, we confirmed the

identity of human and mouse recombinant ACE2 using validated primary antibodies and

conventional Western Blotting (Fig. 7).

Thus, based on these collective data and contrary to our original expectation, we conclude that

DIZE does not enhance activity of ACE2. This conclusion is in contradiction with the premise of

a large body of experimental studies conducted after discovery of DIZE to be an ACE2 activator.

The use of DIZE in these studies was based on the therapeutic benefit attributed to ACE2

activation and enhanced formation of Ang-(1-7) from Ang II. It is important to recognize that

analogues to an enzyme inhibitor, which selectively binds to an enzyme and decreases its

catalytic activity, a small molecule activator would be expected to bind to the enzyme but

increase its catalytic activity. To our knowledge, there are only two reports, in addition to the

original study by Kulemina and Ostrov (2011), showing experimental data with DIZE directly

enhancing activity of ACE2 (Shenoy et al., 2013; LA De Maria et al., 2016). These two papers

19
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

are from the same research group and appear to be related to the team of researchers discovering

small molecule ACE2 activators (Hernandez Prada et al., 2008). On the contrary, several other

studies also looked into confirming activation of ACE2 by DIZE in a limited set of experiments

but failed to document enhanced activity of the peptidase. The first study carried out by Haber

and colleagues (2014) mostly focused on ACE2 activation by another reported ACE2 stimulator,

a xanthenone analog XNT, but also included focused experiments with DIZE. In experiments

involving recombinant ACE2 and synthetic fluorogenic substrate or Ang II, Haber et al. did not

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


document activation of the peptidase by DIZE. Furthermore, the authors did not observe

enhanced activity of ACE2 in mouse and rat kidney lysates in response to DIZE. This was

confirmed by another research group using both recombinant ACE2 and cerebral cortical lysates

(Bennion et al., 2015), although no experimental details were provided. Similarly, Velkoska and

colleagues assessed the direct effect of DIZE on ACE2 activity in rat cardiac (Velkoska et al.,

2016) and kidney cortex (Velkoska et al., 2015) membrane preparations and did not observe

activation of the peptidase. Lastly, another research group evaluated the effect of DIZE on

activity of ACE2 in porcine coronary artery homogenates and failed to document activation

(Raffai et al., 2014). Although, confirmation of ACE2 activation by DIZE was not the primary

goal of these studies and hence only limited number of experiments were carried out, collectively

they support our detailed observations suggesting that this drug does not enhance the catalytic

activity of ACE2. Importantly, several of these studies also included the membrane-bound form

of ACE2 in their experiments, opposite to Kulemina and Ostrov (2011) and the present study that

used the soluble form of the peptidase. Nevertheless, activation of the membrane-bound ACE2

was also not observed in the presence of DIZE.

20
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

As noted above, DIZE has been successfully used in numerous experimental disease models as a

pharmacological treatment, with an understanding that it is an ACE2 activator (Foureaux et al.,

2013; Qi et al., 2013; Rigatto et al., 2013; Shenoy et al., 2013; Bennion et al., 2015; Souza et al.,

2016; Castardeli et al., 2018; Kamel et al., 2018; Evans et al., 2020). A closer look at the

published literature may explain the discrepancy between the lack of ACE2 activation by DIZE

(present and above-discussed studies) and the documented beneficial effects of this drug in

various disease models. One caveat with many of these studies is that activity of ACE2 was

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


simply not measured (Foureaux et al., 2013; Rigatto et al., 2013; Souza et al., 2016; Castardeli et

al., 2018). Another important detail is that many studies administered DIZE over a period of

several days to weeks, and among other effects documented upregulation of ACE2 (at the level

of mRNA and/or protein, e.g. (Foureaux et al., 2013; Qi et al., 2013; Goru et al., 2017; Evans et

al., 2020)). In these cases, higher activity of ACE2 was also documented, which is not surprising

since higher amount of enzyme affords increased catalysis and higher reaction rate within a

certain concentration range (Rashid et al., 2014). In this context, it is important to note that the

primary mechanism of action for the antiprotozoal activity of DIZE is explained by its ability to

intercalate and bind to DNA and RNA duplexes (Pilch et al., 1995). Hence, it is not surprising

that many research groups documented altered expression of ACE2 and other members of the

RAS in response to chronic administration of DIZE in experimental animals. It is important to

recognize that DIZE may also affect the expression of other regulatory proteins involved in

modulation of ACE2. Perhaps, the best example is ADAM17 (a disintegrin and

metalloproteinase 17) which controls shedding of ACE2 ectodomain from the cell membrane

(Xia et al., 2013), and was shown to be downregulated by DIZE (Velkoska et al., 2016; Wang et

al., 2021). ADAM17-mediated regulation of ACE2 appears to be tissues-specific (Pedersen et

21
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

al., 2015) and may be the reason why some studies failed to document DIZE-mediated

upregulation of ACE2 in certain tissues (Velkoska et al., 2016). Furthermore, expression of other

regulatory proteins may also be modulated by DIZE (e.g., proinflammatory cytokines, NADPH

oxidase, extracellular matrix proteins (Rajapaksha et al., 2018)), which offer additional, ACE2-

independent mechanisms to explain the effects of this drug in various pathophysiological

conditions. Lastly, it is important to note that DIZE has been characterized by several research

groups as an acid-sensing ion channel (ASIC) blocker with sub-micromolar potency (Schmidt et

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


al., 2017; Krauson et al., 2018; Lee et al., 2018). This mechanism is likely responsible for both

acute and chronic effects of DIZE, including regulation of blood pressure, inflammation and

ischemic cell death, because of the well-documented role of ASICs in these and other

(patho)physiological conditions (Drummond et al., 2008; Heusser and Pless, 2021).

In conclusion, the experimental observations reported in this study indicate that DIZE does not

enhance catalytic activity of recombinant ACE2 and challenge the widely accepted notion that

this drug is an ACE2 activator. The pharmacological effects of DIZE observed in numerous

publications are likely because of its ability to modulate the expression of various regulatory

proteins and/or blockade of ASICs. Based on this, we urge the scientific community to refrain

from calling DIZE an ACE2 activator. Given that ACE2 is a promising therapeutic target, studies

focusing on identification of small molecule ACE2 activators remain critically important. A

potential lead in this regard could be a recent discovery reporting enhancement of ACE2 activity

by the SARS-CoV-2 spike protein, mediated by binding of its receptor binding domain (RBD)

(Lu and Sun, 2020). Notably, the suggested conformational effects of the spike protein/RBD on

22
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

ACE2 may help to better understand the activation mechanism of ACE2 and design novel small

molecule activators of the peptidase.

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022

23
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

Acknowledgements

We thank Dhaval Patel for his assistance in LC-MS/MS quantification experiments in the initial

phase of this study.

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022

24
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

Author contributions

Participated in research design: Hadi Esfahani and Karamyan.

Conducted experiments: Hadi Esfahani, Jayaraman, and Karamyan.

Performed data analysis: Hadi Esfahani and Karamyan.

Wrote or contributed to the writing of the manuscript: Hadi Esfahani, Jayaraman, and Karamyan.

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022

25
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

References

Al Shoyaib A, Alamri FF, Biggers A, Karamyan ST, Arumugam TV, Ahsan F, Mikelis CM, Al-
Hilal TA and Karamyan VT (2021a) Delayed Exercise-induced Upregulation of
Angiogenic Proteins and Recovery of Motor Function after Photothrombotic Stroke in
Mice. Neuroscience 461:57-71.
Al Shoyaib A, Alamri FF, Syeara N, Jayaraman S, Karamyan ST, Arumugam TV and Karamyan
VT (2021b) The Effect of Histone Deacetylase Inhibitors Panobinostat or Entinostat on
Motor Recovery in Mice After Ischemic Stroke. Neuromolecular Med 23:471-484.
Bader M, Turner AJ and Alenina N (2020) ACE2, a multifunctional protein - from
cardiovascular regulation to COVID-19. Clin Sci (Lond) 134:3229-3232.
Bennion DM, Haltigan EA, Irwin AJ, Donnangelo LL, Regenhardt RW, Pioquinto DJ, Purich

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


DL and Sumners C (2015) Activation of the neuroprotective angiotensin-converting
enzyme 2 in rat ischemic stroke. Hypertension 66:141-148.
Castardeli C, Sartório CL, Pimentel EB, Forechi L and Mill JG (2018) The ACE 2 activator
diminazene aceturate (DIZE) improves left ventricular diastolic dysfunction following
myocardial infarction in rats. Biomedicine & Pharmacotherapy 107:212-218.
Donoghue M, Hsieh F, Baronas E, Godbout K, Gosselin M, Stagliano N, Donovan M, Woolf B,
Robison K and Jeyaseelan R (2000) A novel angiotensin-converting enzyme–related
carboxypeptidase (ACE2) converts angiotensin I to angiotensin 1-9. Circulation research
87:e1-e9.
Drummond HA, Jernigan NL and Grifoni SC (2008) Sensing tension: epithelial sodium
channel/acid-sensing ion channel proteins in cardiovascular homeostasis. Hypertension
51:1265-1271.
Evans CE, Miners JS, Piva G, Willis CL, Heard DM, Kidd EJ, Good MA and Kehoe PG (2020)
ACE2 activation protects against cognitive decline and reduces amyloid pathology in the
Tg2576 mouse model of Alzheimer's disease. Acta Neuropathol 139:485-502.
Foureaux G, Nogueira JC, Nogueira BS, Fulgêncio GO, Menezes GB, Fernandes SO, Cardoso
VN, Fernandes RS, Oliveira GP and Franca JR (2013) Antiglaucomatous effects of the
activation of intrinsic Angiotensin-converting enzyme 2. Investigative ophthalmology &
visual science 54:4296-4306.
Goru SK, Kadakol A, Malek V, Pandey A, Sharma N and Gaikwad AB (2017) Diminazene
aceturate prevents nephropathy by increasing glomerular ACE2 and AT2 receptor
expression in a rat model of type1 diabetes. British journal of pharmacology 174:3118-
3130.
Guy JL, Jackson RM, Acharya KR, Sturrock ED, Hooper NM and Turner AJ (2003)
Angiotensin-converting enzyme-2 (ACE2): comparative modeling of the active site,
specificity requirements, and chloride dependence. Biochemistry 42:13185-13192.
Haber PK, Ye M, Wysocki J, Maier C, Haque SK and Batlle D (2014) Angiotensin-Converting
Enzyme 2–Independent Action of Presumed Angiotensin-Converting Enzyme 2
Activators: Studies In Vivo, Ex Vivo, and In Vitro. Hypertension 63:774-782.
Hernandez Prada JA, Ferreira AJ, Katovich MJ, Shenoy V, Qi Y, Santos RA, Castellano RK,
Lampkins AJ, Gubala V, Ostrov DA and Raizada MK (2008) Structure-based
identification of small-molecule angiotensin-converting enzyme 2 activators as novel
antihypertensive agents. Hypertension 51:1312-1317.

26
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

Heusser SA and Pless SA (2021) Acid-sensing ion channels as potential therapeutic targets.
Trends in pharmacological sciences.
Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, Erichsen S, Schiergens TS,
Herrler G, Wu N-H and Nitsche A (2020) SARS-CoV-2 cell entry depends on ACE2 and
TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell.
Jayaraman S, Al Shoyaib A, Kocot J, Villalba H, Alamri FF, Rashid M, Wangler NJ, Chowdhury
EA, German N, Arumugam TV, Abbruscato TJ and Karamyan VT (2020) Peptidase
neurolysin functions to preserve the brain after ischemic stroke in male mice. J
Neurochem 153:120-137.
Jayaraman S, Kocot J, Esfahani SH, Wangler NJ, Uyar A, Mechref Y, Trippier PC, Abbruscato
TJ, Dickson A, Aihara H, Ostrov DA and Karamyan VT (2021) Identification and
Characterization of Two Structurally Related Dipeptides that Enhance Catalytic
Efficiency of Neurolysin. J Pharmacol Exp Ther 379:191-202.

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


Johnson GD and Ahn K (2000) Development of an internally quenched fluorescent substrate
selective for endothelin-converting enzyme-1. Analytical biochemistry 286:112-118.
Kamel AS, Abdelkader NF, El-Rahman SSA, Emara M, Zaki HF and Khattab MM (2018)
Stimulation of ACE2/ANG (1–7)/Mas axis by diminazene ameliorates Alzheimer’s
disease in the D-galactose-ovariectomized rat model: Role of PI3K/Akt pathway.
Molecular neurobiology 55:8188-8202.
Karamyan VT, Gadepalli R, Rimoldi JM and Speth RC (2009) Brain AT1 angiotensin receptor
subtype binding: importance of peptidase inhibition for identification of angiotensin II as
its endogenous ligand. J Pharmacol Exp Ther 331:170-177.
Karamyan VT and Speth RC (2007) Enzymatic pathways of the brain renin-angiotensin system:
unsolved problems and continuing challenges. Regul Pept 143:15-27.
Krauson AJ, Rooney JG and Carattino MD (2018) Molecular basis of inhibition of acid sensing
ion channel 1A by diminazene. PloS one 13:e0196894.
Kulemina LV and Ostrov DA (2011) Prediction of off-target effects on angiotensin-converting
enzyme 2. Journal of biomolecular screening 16:878-885.
Kuriakose S and Uzonna JE (2014) Diminazene aceturate (Berenil), a new use for an old
compound? International immunopharmacology 21:342-345.
LA De Maria M, D Araújo L, A Fraga-Silva R, AS Pereira L, J Ribeiro H, B Menezes G, Shenoy
V, K Raizada M and J Ferreira A (2016) Anti-hypertensive effects of diminazene
aceturate: an angiotensin-converting enzyme 2 activator in rats. Protein and peptide
letters 23:9-16.
Lee JYP, Saez NJ, Cristofori‐Armstrong B, Anangi R, King GF, Smith MT and Rash LD (2018)
Inhibition of acid‐sensing ion channels by diminazene and APETx2 evoke partial and
highly variable antihyperalgesia in a rat model of inflammatory pain. British journal of
pharmacology 175:2204-2218.
Li W, Moore MJ, Vasilieva N, Sui J, Wong SK, Berne MA, Somasundaran M, Sullivan JL,
Luzuriaga K and Greenough TC (2003) Angiotensin-converting enzyme 2 is a functional
receptor for the SARS coronavirus. Nature 426:450-454.
Lu J and Sun PD (2020) High affinity binding of SARS-CoV-2 spike protein enhances ACE2
carboxypeptidase activity. J Biol Chem 295:18579-18588.
Marquez A, Wysocki J, Pandit J and Batlle D (2021) An update on ACE2 amplification and its
therapeutic potential. Acta Physiol (Oxf) 231:e13513.

27
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

Pedersen KB, Chodavarapu H, Porretta C, Robinson LK and Lazartigues E (2015) Dynamics of


ADAM17-mediated shedding of ACE2 applied to pancreatic islets of male db/db mice.
Endocrinology 156:4411-4425.
Pedersen KB, Sriramula S, Chhabra KH, Xia H and Lazartigues E (2011) Species-specific
inhibitor sensitivity of angiotensin-converting enzyme 2 (ACE2) and its implication for
ACE2 activity assays. American Journal of Physiology-Regulatory, Integrative and
Comparative Physiology 301:R1293-R1299.
Pilch DS, Kirolos MA, Liu X, Plum GE and Breslauer KJ (1995) Berenil [1, 3-bis (4'-
amidinophenyl) triazene] binding to DNA duplexes and to a RNA duplex: evidence for
both intercalative and minor groove binding properties. Biochemistry 34:9962-9976.
Qi Y, Zhang J, Cole-Jeffrey CT, Shenoy V, Espejo A, Hanna M, Song C, Pepine CJ, Katovich
MJ and Raizada MK (2013) Diminazene aceturate enhances angiotensin-converting
enzyme 2 activity and attenuates ischemia-induced cardiac pathophysiology.

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


Hypertension 62:746-752.
Raffai G, Khang G and Vanhoutte PM (2014) Angiotensin-(1-7) augments endothelium-
dependent relaxations of porcine coronary arteries to bradykinin by inhibiting
angiotensin-converting enzyme 1. Journal of cardiovascular pharmacology 63:453-460.
Rahman MS, Kumari S, Esfahani SH, Nozohouri S, Jayaraman S, Kinarivala N, Kocot J, Baez
A, Farris D, Abbruscato TJ, Karamyan VT and Trippier PC (2021) Discovery of First-in-
Class Peptidomimetic Neurolysin Activators Possessing Enhanced Brain Penetration and
Stability. J Med Chem 64:12705-12722.
Rajapaksha IG, Mak KY, Huang P, Burrell LM, Angus PW and Herath CB (2018) The small
molecule drug diminazene aceturate inhibits liver injury and biliary fibrosis in mice.
Scientific reports 8:10175.
Rashid M, Wangler NJ, Yang L, Shah K, Arumugam TV, Abbruscato TJ and Karamyan VT
(2014) Functional upregulation of endopeptidase neurolysin during post-acute and early
recovery phases of experimental stroke in mouse brain. Journal of neurochemistry
129:179-189.
Rigatto K, Casali KR, Shenoy V, Katovich MJ and Raizada MK (2013) Diminazene aceturate
improves autonomic modulation in pulmonary hypertension. Eur J Pharmacol 713:89-
93.
Schmidt A, Rossetti G, Joussen S and Gründer S (2017) Diminazene is a slow pore blocker of
acid-sensing ion channel 1a (ASIC1a). Molecular pharmacology 92:665-675.
Shenoy V, Gjymishka A, Jarajapu YP, Qi Y, Afzal A, Rigatto K, Ferreira AJ, Fraga-Silva RA,
Kearns P and Douglas JY (2013) Diminazene attenuates pulmonary hypertension and
improves angiogenic progenitor cell functions in experimental models. American journal
of respiratory and critical care medicine 187:648-657.
Shrimpton CN, Glucksman MJ, Lew RA, Tullai JW, Margulies EH, Roberts JL and Smith AI
(1997) Thiol activation of endopeptidase EC 3.4. 24.15 A novel mechanism for the
regulation of catalytic activity. Journal of Biological Chemistry 272:17395-17399.
Souza LKM, Nicolau LA, Sousa NA, Araújo TS, Sousa FBM, Costa DS, Souza FM, Pacífico
DM, Martins CS and Silva RO (2016) Diminazene aceturate, an angiotensin-converting
enzyme II activator, prevents gastric mucosal damage in mice: Role of the angiotensin-
(1–7)/Mas receptor axis. Biochemical pharmacology 112:50-59.

28
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

Tipnis SR, Hooper NM, Hyde R, Karran E, Christie G and Turner AJ (2000) A human homolog
of angiotensin-converting enzyme cloning and functional expression as a captopril-
insensitive carboxypeptidase. Journal of Biological Chemistry 275:33238-33243.
Velkoska E, Patel SK, Griggs K and Burrell LM (2016) Diminazene aceturate improves cardiac
fibrosis and diastolic dysfunction in rats with kidney disease. PloS one 11:e0161760.
Velkoska E, Patel SK, Griggs K, Pickering RJ, Tikellis C and Burrell LM (2015) Short-term
treatment with diminazene aceturate ameliorates the reduction in kidney ACE2 activity in
rats with subtotal nephrectomy. PLoS One 10:e0118758.
Vickers C, Hales P, Kaushik V, Dick L, Gavin J, Tang J, Godbout K, Parsons T, Baronas E,
Hsieh F, Acton S, Patane M, Nichols A and Tummino P (2002) Hydrolysis of biological
peptides by human angiotensin-converting enzyme-related carboxypeptidase. J Biol
Chem 277:14838-14843.
Wang L-F, Sun Y-Y, Pan Q, Yin Y-Q, Tian X-M, Liu Y, Bu T, Zhang Q, Wang Y-A and Zhao J

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


(2021) Diminazen Aceturate Protects Pulmonary Ischemia-Reperfusion Injury via
Inhibition of ADAM17-Mediated Angiotensin-Converting Enzyme 2 Shedding. Frontiers
in Pharmacology:2700.
Wangler NJ, Jayaraman S, Zhu R, Mechref Y, Abbruscato TJ, Bickel U and Karamyan VT
(2016) Preparation and preliminary characterization of recombinant neurolysin for in
vivo studies. Journal of biotechnology 234:105-115.
Wösten-van Asperen R, Lutter R, Haitsma J, Merkus M, van Woensel J, Van Der Loos C,
Florquin S, Lachmann B and Bos A (2008) ACE mediates ventilator-induced lung injury
in rats via angiotensin II but not bradykinin. European Respiratory Journal 31:363-371.
Xia H, Sriramula S, Chhabra KH and Lazartigues E (2013) Brain angiotensin-converting enzyme
type 2 shedding contributes to the development of neurogenic hypertension. Circulation
research 113:1087-1096.

29
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

Footnotes

This work was in part supported by a research grant from the NINDS [1R01NS106879]. No

author has an actual or perceived conflict of interest with the contents of this article.

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022

30
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

Figure legends

Figure 1. Effect of DIZE on activity of human and mouse recombinant ACE2 in Tris

buffer. Panels a and b: representative reaction progress curves of fluorogenic substrate

hydrolysis (10 µM) by recombinant human and mouse ACE2 in the presence of different

concentrations of DIZE in Tris HCl assay buffer used by Kulemina and Ostrov (2011). Panels c

and d: concentration-dependent effect of DIZE on hydrolysis of the fluorogenic substrate under

the same experimental conditions (mean ± SD, n = 6 independent experiments with duplicate

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


samples for each condition). Note that the initial velocity of hydrolysis in the absence of DIZE

corresponds to 100% on the vertical axis (basal activity). Panels e and f: representative,

concentration-dependent effect of DIZE on fluorescence signal of Mca-Pro-Leu-OH, under the

same assay conditions. The only difference was presence of McaPro-Leu-OH, instead of the

fluorogenic substrate, in the assay at 2 µM final concentration. Each data point represents the

average fluorescence signal measured every minute for a 10 min period.

Figure 2. Effect of DIZE on activity of peptidases in aCSF. All panels document

concentration-dependent effect of DIZE on hydrolysis of a respective fluorogenic substrate under

the same experimental conditions described for Fig. 1, with the exception that aCSF was used as

assay buffer (mean ± SD, n = 6 independent experiments with duplicate samples for each

condition): Mca-Ala-Pro-Lys (Dnp) at 10 µM for human and mouse ACE2, Mca-Arg-Pro-Pro-

Gly-Phe-Ser-Ala-Phe-Lys(Dnp)-OH at 10 µM for neprilysin (NEP) and angiotensin converting

enzyme (ACE), Mca-Pro-Leu-Gly-Pro-D-Lys(DNP)-OH at 15 µM for neurolysin (Nln) and

thimet oligopeptidase (TOP). Note that the initial velocity of hydrolysis in the absence of DIZE

corresponds to 100% on the vertical axis (basal activity).

31
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

Figure 3. The effect of DIZE on catalytic efficiency of human and mouse recombinant

ACE2. Hydrolysis of different concentrations of fluorogenic substrate (QFS) by human and

mouse recombinant ACE2 in the absence or presence of DIZE (10 and 50 µM) is presented

(mean ± SD, n = 6 independent experiments with duplicate samples for each condition, FLU –

fluorescence unit).

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


Figure 4. The effect of DIZE on hydrolysis of Ang II by ACE2. Human or mouse recombinant

ACE2 (0.3 and 0.5 nM, respectively) was incubated with Ang II (20 µM) in the absence or

presence of DIZE (10 or 50 µM). Relative amounts of Ang-(1-7) and Ang II were documented

by mass spectrometry analysis (n = 6 independent experiments with duplicate samples for each

condition; p > 0.05 for respective ACE2 alone vs. ACE2 with either concentration of DIZE; the

black line within the scattered dots indicates the mean). Analyte denotes Ang II or Ang-(1-7),

whereas IS denotes internal standard.

Figure 5. Inhibition profile of ACE2 by DX600 and MLN4760. Concentration-dependent

effect of DX600 and MLN4760 on hydrolysis of the fluorogenic substrate by human (a) and

mouse (b) recombinant ACE2 (mean ± SD, n = 6 independent experiments with duplicate

samples for each condition). Note that the initial velocity of hydrolysis in the absence of the

inhibitors corresponds to 100% on the vertical axis (basal activity).

Figure 6. Influence of NaCl on activity of ACE2. Concentration-dependent effect of NaCl on

hydrolysis of the fluorogenic substrate by human (a) and mouse (b) recombinant ACE2 (n = 6

32
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

independent experiments with duplicate samples for each condition; FLU – fluorescence unit;

**p<0.01, ***p<0.001 in comparison to respective ACE2 activity in the absence of NaCl; the

black line within the scattered dots indicates the mean).

Figure 7. Identification of ACE2 by Western Blotting. Representative immunoblotting

experiment documenting identity of the commercial, recombinant human (h) and mouse (m)

ACE2 used in this study. Human (20 and 40 ng) and mouse (40 and 80 ng) ACE2, and human

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


ACE (negative control, 20 and 40 ng) were subjected to SDS-PAGE analysis followed by

conventional Western Blotting to first detect ACE2 and then (after stripping the anti-ACE2

antibody), ACE. Migration of molecular weight standards (25 to 250 kDa) are presented on the

left side of the image.

33
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

Tables

Table 1. Calculated Vmax and Km values for experiments presented in Figure 3.

Vmax Km Vmax/Km Fold change

791.2 19.4
human ACE2 40.7 ---
(745.3 – 837) (16.2 – 22.6)

808.2 21.6
hACE2 + 10µM DIZE 37.4 0.92
(742 – 874.4) (16.7 – 26.5)

595.8*** 12.5*
hACE2 + 50µM DIZE 47.8 1.17
(549.3 – 642.4) (9.3 – 15.6)

560.5 9.8

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


mouse ACE2 56.7 ---
(525.9 – 595.1) (7.7 – 12)

509.2 9.6
mACE2 + 10µM DIZE 52.8 0.93
(460.5 – 557.9) (6.4 – 12.8)

418.3*** 8.4
mACE2 + 50µM DIZE 49.9 0.88
(378.5 – 458.2) (5.5 – 11.2)

Vmax units are in FLU/ng protein per min (FLU – fluorescence unit), Km units are in
µM. Data are presented as means together with 95% confidence intervals in parenthesis
(n = 6 independent experiments with duplicate samples for each condition; *p<0.05,
***p<0.001 in comparison to respective ACE2 values).

34
Figure 1.
4000 4000
Fluorescence units (FLU)

Fluorescence units (FLU)


human ACE2 A mouse ACE2 B
0µM 0µM
3000 0.1µM 3000 0.1µM
0.3µM 0.3µM
1µM 1µM

2000 2000

3µM 3µM
1000 10µM 1000 10µM
30µM 30µM
100µM 100µM

0 0
0 5 10 15 20 0 5 10 15 20
Time (min) Time (min)

150 150
human ACE2 C mouse ACE2 D
125 125

100 100
% V0
% V0

75 75

50 50

25 25

0 0
-7 -6 -5 -4 -7 -6 -5 -4
[DIZE], log M [DIZE], log M

4000 4000
Fluorescence units (FLU)

Fluorescence units (FLU)

fluorescence quenching E fluorescence quenching F


human ACE2 mouse ACE2
3000 3000

2000 2000

1000 1000

0 0
-13 -7 -6 -5 -4 -13 -7 -6 -5 -4
[DIZE], log M [DIZE], log M
Figure 2.
150 150
human ACE2 A mouse ACE2 B
125 125

100 100
% V0

% V0
75 75

50 50

25 25

0 0
-7 -6 -5 -4 -7 -6 -5 -4
[DIZE], log M [DIZE], log M

150 150
human ACE C human Nln D
125 125

100 100
% V0

% V0

75 75

50 50

25 25

0 0
-7 -6 -5 -4 -7 -6 -5 -4
[DIZE], log M [DIZE], log M

150 150
human TOP E human NEP F
125 125

100 100
% V0

% V0

75 75

50 50

25 25

0 0
-7 -6 -5 -4 -7 -6 -5 -4
[DIZE], log M [DIZE], log M
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


Figure 3.

800 human ACE2 A 800 mouse ACE2 B


(FLU/ng protein per min)

(FLU/ng protein per min)


600 600
Vo

Vo

400 400

200 0 μM 200 0 μM
10 μM 10 μM
50μM 50μM
0 0
0 25 50 75 100 0 25 50 75 100
QFS (μM) QFS (μM)
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
Figure 4. This article has not been copyedited and formatted. The final version may differ from this version.
2.0

Ang-(1-7), relative amount


A

(peak area analyte/IS)


1.5

1.0

0.5

0.0
0 0 10 50 0 10 50

no ACE2 DIZE (µM)


human ACE2 mouse ACE2

2.0
B
Ang II, relative amount
(peak area analyte/IS)

1.5

1.0

0.5

0.0
0 0 10 50 0 10 50
DIZE (µM)
no ACE2

human ACE2 mouse ACE2

2.0
C
(ratio of relative amounts)
Ang-(1-7) / Ang II

1.5

1.0

0.5

0.0
0 0 10 50 0 10 50
DIZE (µM)
no ACE2

human ACE2 mouse ACE2


JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

Figure 5.

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


125 125
human ACE2 A mouse ACE2 B
100 100

75 75
% V0
% V0

50 50

25 25
DX600 DX600
0 MLN 4760 0 MLN 4760

-11 -10 -9 -8 -7 -6 -11 -10 -9 -8 -7 -6


[inhibitor], log M [inhibitor], log M
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


Figure 6.

human ACE2 A mouse ACE2 B


(FLU/ng protein per min)
(FLU/ng protein per min)

800 800
***
*** **
600 600
Vo
Vo

400 *** 400 ***

200 200

0 0
0 0.1 0.5 1 0 0.1 0.5 1
NaCl (M) NaCl (M)
JPET Fast Forward. Published on September 2, 2022 as DOI: 10.1124/jpet.122.001339
This article has not been copyedited and formatted. The final version may differ from this version.

Downloaded from jpet.aspetjournals.org at ASPET Journals on September 19, 2022


Figure 7.

You might also like