ELA-11 Protects The Heart Against Oxidative Stress
ELA-11 Protects The Heart Against Oxidative Stress
ELA-11 Protects The Heart Against Oxidative Stress
through a 180-mesh filters. The cells were resuspended in 10 ml Lactate dehydrogenase level detection
DMEM containing 10% horse serum (HS, Gibco, United States)
and incubated in a 10 cm2 dish for 1.5 h. The cell suspension was Levels of lactate dehydrogenase (LDH) released were
removed, and the cells (5–6 × 105 cells per dish) were inoculated detected in serum using an LDH release assay kit according to
into a new dish as previously described. When cell adhered to the manufacturer’s protocol.
about 80%, 1uM ELA-11 was added to the culture media. After
12 h of co-incubation with DOX, experimental verification was
carried out. Terminal-deoxynucleotidyl transferase-
mediated dUTP-biotin nick end-labeling
staining assay
Animals
The rate of apoptosis can be was detected by a TUNEL
Male C57BL/6J mice (6–8 weeks of age, 20–22 g) were obtained staining kit. The cells were seeded (1 × 105 cells per well) in 6-well
from the Model Animal Research Center of Nanjing University dishes. After the treatments described above were performed, the
(Nanjing, Jiangsu, China), and all procedures used were approved by cells were washed once with phosphate-buffered saline (PBS) and
the ethical committee of Nanjing Medical University. All animals fixed with 4% paraformaldehyde. Apoptotic cells were visualized
were housed at 20–25°C and 50%–70% relative humidity. The with TUNEL staining according to the manufacturer’s protocol.
experimental mice were randomly divided into four groups. TUNEL fluorescence intensity/DAPI fluorescence density was
Before injected with 5 mg/kg DOX by intraperitoneal injection used to calculate the percentage of positive cells, and the density
for five consecutive weeks, 10 mg/kg ELA-11 was injected to was evaluated using ImageJ software 1.26 (Wayne Rasband,
mice through veil tail for 7 days, after which electrocardiograms National Institutes of Health, Bethesda, MD, United States).
were obtained and the mice were sacrificed. The mice were treated
according to the experimental requirements. All animal experiments
complied with the Guide for the Care and Use of Laboratory Tetrechloro-tetraethylbenzimidazol
Animals published by the National Institutes of Health (NIH carbocyanine iodide assay
Publications No. 85-23, revised 1996).
The mitochondrial membrane potential was measured by a
mitochondrial membrane potential assay kit with JC-1 according
Cell-in-cell experiment to the manufacturer’s instructions. The cells were cultured in
serum-free DMEM containing (×1) JC-1 staining working fluid
We chemically synthesized FITC-labeled, incubated cells at 37°C for 20 min. Then, the cells were washed twice with JC-1
with ELA-11 for 1 h at 37°C and 5% CO2 atmosphere. The buffer, after which 2 ml DMEM was added, and then the cells
localization of ELA-11 was observed by fluorescence microscopy. were photographed with a fluorescence microscope (BX61;
Olympus Corporation, Tokyo, Japan). The JC-1 density was
assessed by ImageJ software and calculated upon
Cell counting kit-8 assay normalization to the control.
FIGURE 1
Function of ELA-11 in doxorubicin-injured model. (A) The biological characteristics of ELA. (B) Schematic map for DOX-induced mice cardiac
injury model. (C) The body weight of mice in four groups during the experiment. (D) The representative echocardiograms of DOX-induced cardiac
injured group. (E) EF value for echocardiography. (F) FS value for echocardiography. (G) LVEDs value for echocardiography. ***p < 0.001 and ### p <
0.001, one-way ANOVA, N = 5 mice per group.
Fisher Scientific). Protein samples of the same mass were standard substance was prepared and diluted according to the
separated on 10% SDS-PAGE gels and transferred to instructions, the concentration of each well was 600, 400, 200,
nitrocellulose membranes (Millipore, Billerica, MA, 100, and 50 U/L, respectively. The tested samples were added to
United States), which were blocked with 5% skim milk and the plate and mixed, then incubated at 37°C for 30 min. Wash
then incubated with specific primary antibodies. Caspase-3 plates, add enzyme and chromogenic reaction were proceeded
(14220T, 1:1000), PARP (9532T, 1:1000), β-actin (3700S, 1: according to the instructions. After incubating at 37°C for
1000), GAPDH (5174T, 1:1000), AKT (4685S, 1:1000), p-AKT 15 min, the absorbance of each well was measured at 450 nm
(4060T, 1:2000), PI3K (4249T, 1:1000), ERK (4695T, 1:1000) and wavelength.
p-ERK (4370T, 1:1000) were purchased from Cell signaling
Technology. The anti-rabbit (SA00001-2, 1:10000), PI3K
(20584-1-AP, 1:1000) and anti-mouse (SA00001-1, 1:10000) Echocardiography
secondary antibody were purchase from Proteintech.
FluorChem M system was used to quantify the positive bands Post DOX and ELA-11 administration, all the mice were
representing proteins involved in the orchestrated immune subjected to M-mode echocardiography to assess heart
responses (ProteinSimple, San Jose, CA, United States). function. All animal experiments complied with the Guide
for the Care and Use of Laboratory Animals published by the
National Institutes of Health (NIH Publications No. 85-23,
Malondialdehyde, superoxide dismutase, revised 1996).
and glutathione peroxidase assay
Cellular oxidative stress was determined by detecting the Enzyme-linked immunosorbent assay
state of intracellular oxidation and reduction. Lipid peroxidation
Malondialdehyde (MDA) assay (Beyotime, Nanjing, China) and Serum Creatine kinase isoenzyme (CKMB) and B-type
total superoxide dismutase assay (Beyotime, Nanjing, China) natriuretic peptide (BNP) level in mice heart tissue was
were measured by following the manufacturer’s instructions. determined by a commercially available Enzyme-linked
glutathione peroxidase (GSH-Px) assay kit was purchased immunosorbent assay (ELISA) kit (Mlbio, Shanghai, China)
from Elabscience. The cell supernatant was collected, and the according to the manufacturer’s instructions.
FIGURE 2
ELA-11 protects doxorubicin-injured heart in vivo. (A) Serum CKMB release in DOX-injured model. (B) Serum BNP release in DOX-injured
model. (C) Representative photographs of HE staining. (D) Representative photographs of Masson staining. (E,F) Representative photographs of WGA
staining and its quantitative data. (G,H) Representative photographs of TUNEL staining and its quantitative data. ***p < 0.001 and ### p < 0.001, one-
way ANOVA, N = 5 mice per group.
Hematoxylin & Eosin (H&E) Staining and ponceau acid fuchsin solution was used for 5–10 min after
washing. Soaked with 2% glacial acetic acid solution for 30 s, and
Obtained the cardiac tissues and fixed them in 4% differentiate with 1% phosphomolybdate solution for 3–5 min.
paraformaldehyde for 7 days after the blood in the heart Then, the slices were dyed with aniline blue for 5 min, and
cavity was pumped out. The tissue was transparent with bathed again with 0.2% glacial acetic acid solution for 30 s.
ethanol and xylene. Soak the transparent tissue into melted Finally, the slices were sealed with 95% alcohol, anhydrous
paraffin for embedding. After cooling and solidification, they alcohol, xylene transparent and neutral gum.
were cut into five micron slices and placed in hot water to flatten
and then pasted onto slides and dried in a 45°C incubator. Before
staining, paraffin was removed again, and Hematoxylin and eosin Wheat germ agglutinin staining
dyes were respectively used for staining, which made the nucleus
and intracellular ribosomes stained blue and purple by Obtained and sectioned tissue according to the above
Hematoxylin (H). The cytoplasm is stained red or reddish method. Placed the section in the antigen repair buffer of
with Eosin (E). pH 8.0, then the section was decolorized with PBS after
cooled naturally. A histochemical pen was used to draw
circles around the tissues and Wheat germ agglutinin (WGA)
Masson staining staining buffer was added into the circles, and the cells were
incubated for 30 min at 37°C. The nucleus was stained with
Obtained and sectioned tissue according to the above method. DAPI. Sealed the section, then collected the images under
Weigert sapwood semen was used for staining nuclear for 5–10 min, fluorescence microscope.
FIGURE 3
ELA-11 attenuates doxorubicin-induced injury in rat primary cardiomyocytes. (A) The location of ELA-11 in rat primary cardiomyocytes. (B)
Trypan blue assay for cell death with different concentration of DOX with different duration (N = 9 per group). (C) The cell viability of 1 uM DOX in
different duration (N = 9 per group). (D) Cell viability of ELA-11 with different concentration for 12 h (N = 9 per group). (E) Cell viability in DOX-induced
cardiomyocyte model (N = 9 per group). (F) LDH release in DOX-induced cardiomyocyte model (N = 9 per group). ***p < 0.001 and ### p <
10.001, one-way ANOVA.
FIGURE 4
ELA-11 inhibits DOX-induced cardiac injury through oxidative stress -induced apoptosis. (A) Representative photographs of TUNEL staining in
DOX- induced cardiomyocyte injury (N = 9 per group). (B) Representative photographs of JC-1 in DOX-induced cardiomyocyte injury. (C) The
expression of PARP and caspase-3 in DOX-induced cardio-myocyte injury by western blot. (D) Representative photographs of ROS in DOX-induced
cardio-myocyte injury. (E) Detection of MDA production, SOD activity and GSH-PX release in DOX-induced cardiomyocyte injury (N = 9 per
group). ***p < 0.001 and ### p < 0.001, one-way ANOVA.
significantly in DOX-treated group compared with the Scr group, was no statistic difference between the two groups (Figure 3C).
but ELA-11 significantly rescued cardiomyocyte area To figure out whether ELA-11 could affect cell survival, we
(Figures 2F,G). TUNEL staining showed a significant increase cultured ELA-11 with different duration times (0, 6, 12, and
in the apoptosis rate in the myocardium in the DOX + Scr group, 24 h) and concentrations (0.1, 0.5, 1, 2, and 5 uM). Then trypan
and it generally decreased to the level in DOX mice administered blue dyeing results demonstrated that 0.5 uM ELA-11 decreased
with ELA-11 (Figures 2H,I). These results demonstrated that cell death at 24 h, 1 uM, and 5 uM ELA-11 both could decrease
ELA-11 might have a protective role in DOX-induced cardiac cell death at 12 h. But there was no significant difference among
injury. the groups (Figure 3D). Next, we cultured 1 uM ELA-11 before
DOX treatment for 12 h and found ELA-11 could significantly
increase cell viability compared with DOX + Scr group
ELA-11 attenuates doxorubicin-induced (Figure 3E). And LDH results indicated that ELA-11 inhibited
injury in rat primary cardiomyocytes DOX-induced cardiotoxicity (Figure 3F). Our study revealed that
ELA-11 could attenuate DOX-induced injury in rat primary
To determine the effect of ELA-11 in vitro, we first cultured cardiomyocytes.
rat primary cardiomyocytes with ELA-11, we found that ELA-11
could enter into the cytoplasm (Figure 3A). To figure out the
optimal concentration of DOX, we incubated DOX with different ELA-11 inhibits doxorubicin-induced
concentrations (0.1, 0.5, 1, 2, and 5 uM) for 12 h, CCK-8 results cardiac injury through oxidative stress-
demonstrated that 0.5 uM DOX could reduce cell viability, but 1, induced apoptosis
2, and 5 uM decreased cell viability more significantly and there
were no difference among 1, 2 and 5 uM DOX (Figure 3B). Then Based on the effect of ELA-11 in vivo, we further detected the
CCK-8 results manifested that 1 uM DOX cultured for 12 and apoptosis level by TUNEL, the results revealed that the number
24 h could effectively reduce the cell survival rate, however there of apoptotic cells was increased in the DOX-induced group but
FIGURE 5
ELA-11 protected cardiomyocytes against oxidative stress-induced apoptosis in CoCl2 model. (A) Cell viability in CoCl2 -induced
cardiomyocyte model. (B) LDH release in CoCl2 -induced cardiomyocyte oxidative stress injury (N = 9 per group). (C,D) The expression of PARP and
caspase-3 in CoCl2 -induced cardiomyocyte oxidative stress injury by western blot. (E) Representative photographs of ROS production in CoCl2
-induced cardiomyocyte oxidative stress injury. (F–H) Detection of MDA production, SOD activity and GSH -PX release in CoCl2 -induced
cardiomyocyte oxidative stress injury (N = 9 per group). ***p < 0.001 and ### p < 0.001, one-way ANOVA.
reduced after ELA-11 co-treatment (Figure 4A). Furthermore, inhibit CoCl2-induced cardiac apoptosis (Figures 5C,D).
DOX reduced mitochondrial membrane potential compared Furthermore, ELA-11 inhibited ROS production with CoCl2
with scr group, but ELA-11 elevated mitochondrial membrane treatment (Figure 5E). And ELA-11 inhibited superoxide
potential compared with DOX treatment (Figure 4B). The production and peroxidase activity (Figures 5F–H). Our
western blot results revealed that cleaved caspase-3 and PARP results further proved ELA-11 protected cardiac injury in
were activated by DOX, but ELA-11 reduced the elevated CoCl2 model mediated by oxidative stress-induced apoptosis.
expression of cleaved caspase-3 and PARP (Figure 4C). Next,
ROS release results indicated that ELA-11 attenuated DOX-
induced free radical production (Figure 4D). Furthermore, we ELA-11 attenuated cardiac injury through
evaluated cellular oxidative stress levels by MDA production, ERK/MAPK and PI3K/AKT signaling
SOD activity and GSH-Px content. Our results demonstrated pathway
that DOX increased MDA production and reduced SOD activity
and GSH-Pxproduction, but ELA-11 inhibited MDA production, We further verified the possible signaling pathway of ELA-11
promoted SOD activity and GSH-Px production (Figure 4E). in rat primary cardiomyocytes by Western blot. We first
Therefore, ELA-11 could inhibit DOX-induced injury by determined the expression of p38, JNK and ERK, however,
attenuating oxidative stress-induced apoptosis. the result showed that there was no expression of JNK and
p38 (data not shown), but phosphorylated ERK was down-
regulated by DOX and ERK protein phosphorylation was
ELA-11 protected cardiomyocytes against activated upon ELA-11 treatment compared with DOX
oxidative stress-induced apoptosis in (Figures 6A,B). Next, we found that DOX also down-regulated
CoCl2 model the expression of phosphorylated AKT and PI3K, but ELA-11
activated phosphorylation of AKT and PI3K protein expression
To validate the role of ELA-11 in another oxidative stress (Figures 6C–E). When CoCl2 was blunted, the phosphorylation
model, we further constructed chemical ischemic model in rat of ERK, AKT and PI3K protein expression was downregulated.
primary cardiomyocytes. Our results indicated that ELA-11 With intervention of ELA-11, ELA-11 activated the expression of
protected cardiomyocytes from CoCl2-induced injury (Figures ERK, AKT and PI3K phosphorylated proteins after CoCl2
5A,B). Then western blot evidence proved that ELA-11 could treatment in rat primary cardiomyocytes (Figures 6F–J). These
FIGURE 6
ELA-11 attenuated cardiac injury through ERK/MAPK and PI3K/AKT signaling pathway. (A) The expression of ERK in DOX-induced cardiac injury
by western blot. (B) The quantitative data for western blot. (C–E) The expression of PI3K and AKT in DOX-induced cardiac injury by western blot and
its quantitative data. (F,G) Western blot analyzed the expression the expression of ERK in CoCl2 -induced cardiac injury and its quantitative data.
(H–J) Western blot analyzed the expression the expression of PI3K and AKT in CoCl2 -induced cardiac injury and its quantitative data. ***p <
0.001 and ### p < 0.001, one-way ANOVA.
results revealed that ELA-11 protected cardiomyocytes from and western blot results demonstrated that
apoptosis through the ERK/MAPK and PI3K/AKT signaling ML221 significantly decreased the phosphorylation of AKT,
pathways. PI3K, and ERK proteins compared with ELA-11, when ELA-
11 and ML221 co-incubation with DOX, the ability of ELA-11 to
activate phosphorylated ERK, PI3K and AKT was inhibited
ELA-11 protects cardiomyocytes by (Figures 7C–G). Furthermore, we found that ML221 could
binding apelin receptor also inhibit the effect of ELA-11 in CoCl2-induced apoptosi s
(Figures 7H,I). The phosphorylation of AKT, PI3K and ERK
4- oxo-6-((pyrimidin-2-ylthio) methyl)-4H-pyran-3-yl4- proteins was significantly up-regulated after co-treatment with
nitrobenzoate (ML221) is the first reported APJ antagonist ML221 and ELA-11 in CoCl2 model (Figures 7J,K,L–N). These
which exerts antagonistic effect mainly by inhibiting cAMP results suggested that ELA-11 inhibited apoptosis by binding
and recruiting β-arrestin by ELA-11. Report has shown that to APJ. Finally, we drew a pattern diagram of ELA-11 for
ML221 has potential to attenuate the activation and signaling of inhibiting apoptosis and oxidative stress injury in
the APJ receptor and reduce elabela-induced microvascular cardiomyocytes (Figures 8).
endothelial cell proliferation (Godoy-Parejo et al., 2019). To
determine the role of ML221, we used to detect its interaction
with ELA-11 by Western blot. We found that ML221 inhibited Discussion
the effect of ELA-11 on cardiomyocyte apoptosis after DOX
administration (Figures 7A,B). We wondered whether Our current study revealed that ELA-11 has a protective
ML221 was involved in the ERK/MAPK and PI3K/AKT effect on apoptosis and attenuates DOX-induced cardiotoxicity
signaling pathways when the cells were treated with DOX, in vitro and in vivo. Furthermore, we demonstrated that ELA-11
FIGURE 7
ELA-11 protects cardiomyocytes by binding APJ. (A,B) Western blot result of PARP and caspase-3 with ML221 treatment in DOX-induced group
and its quantiied data. (C,D) Western blot result of ERK with ML221 treatment in DOX -induced group and its quantiied data. (E–G) Western blot and
quantiied data for PI3K and AKT in DOX-induced group. (H,I) The expression of PARP and casapse-3 in CoCl2 -induced group by western blot and
quantiied. (J,K) The expression of ERK in CoCl2 -induced group by western blot and quantiied. (L–N) The expression of PI3K and AKT in CoCl2
-induced group with quantiied data. ***p < 0.001, and ### p < 0.001, one-way ANOVA.
attenuated oxidative stress-induced apoptosis through ERK/ of disease monitoring (Ishimaru et al., 2017). In addition,
MAPK and PI3K/AKT signaling pathways by targeting endocrine-derived peptides play a dispensable role in different
APJ. ERK/MAPK and PI3K/AKT signaling pathways is pathological processes and engage in organ crosstalk. For
regarded as a promising molecular mechanism in apoptosis, instance, a vasoactive intestinal polypeptide of 28 amino acids
proliferation and differentiation. Our findings provide is released by intestinal neurons (Reginauld et al., 2019). Its level
evidence of the molecular mechanism by which the ELA-11 of variation is related to a variety of human diseases. It is worth
peptide inhibits oxidative stress-induced apoptosis, indicating its observing that some long peptides can be cleaved into smaller
potential use for DOX-induced cardiotoxicity. peptides that have more consequential functions than those of
Many known bioactive substances secreted by heart, longer peptides.
including ANP, have been indicated to be involved in the Because of the cardiac toxicity caused by DOX, its clinical
regulation of cardiovascular disease and sensitive as indicators application is severely limited (Sun et al., 2019). Apoptosis and
References
Ahmad, F., Lal, H., Zhou, J., Vagnozzi, R. J., Yu, J. E., Shang, X., et al. (2014). pathways. Antimicrob. Agents Chemother. 58 (7), 4075–4085. doi:10.1128/AAC.
Cardiomyocyte-specific deletion of Gsk3α mitigates post-myocardial infarction 00070-14
remodeling, contractile dysfunction, and heart failure. J. Am. Coll. Cardiol. 64 (7),
Dai, M., Cui, P., Yu, M., Han, J., Li, H., and Xiu, R. (2008). Melatonin modulates
696–706. doi:10.1016/j.jacc.2014.04.068
the expression of VEGF and HIF-1 alpha induced by CoCl2 in cultured cancer cells.
Ahmad, F., Singh, A. P., Tomar, D., Rahmani, M., Zhang, Q., Woodgett, J. R., et al. J. Pineal Res. 44 (2), 121–126. doi:10.1111/j.1600-079X.2007.00498.x
(2019). Cardiomyocyte-GSK-3α promotes mPTP opening and heart failure in mice
Eid, B. G., El-Shitany, N. A. E., and Neamatallah, T. (2021). Trimetazidine
with chronic pressure overload. J. Mol. Cell. Cardiol. 130, 65–75. doi:10.1016/j.
improved adriamycin-induced cardiomyopathy by downregulating TNF-α, BAX,
yjmcc.2019.03.020
and VEGF immunoexpression via an antioxidant mechanism. Environ. Toxicol. 36,
Ahmad, F., and Woodgett, J. R. (2020). Emerging roles of GSK-3α in 1217–1225. doi:10.1002/tox.23120
pathophysiology: Emphasis on cardio-metabolic disorders. Biochim. Biophys.
Godoy-Parejo, C., Deng, C., Liu, W., and Chen, G. (2019). Insulin stimulates
Acta. Mol. Cell. Res. 1867 (2), 118616. doi:10.1016/j.bbamcr.2019.118616
PI3K/AKT and cell adhesion to promote the survival of individualized
Brady, D., Grapputo, A., Romoli, O., and Sandrelli, F. (2019). Insect cecropins, human embryonic stem cells. Stem Cells 37 (8), 1030–1041. doi:10.1002/
antimicrobial peptides with potential therapeutic applications. Int. J. Mol. Sci. 20 stem.3026
(23), 5862. doi:10.3390/ijms20235862
Govender, J., Loos, B., Marais, E., and Engelbrecht, A. M. (2014). Mitochondrial
Carrasco, R., Castillo, R. L., Gormaz, J. G., Carrillo, M., and Thavendiranathan, P. catastrophe during doxorubicin-induced cardiotoxicity: A review of the protective
(2021). Role of oxidative stress in the mechanisms of anthracycline-induced role of melatonin. J. Pineal Res. 57 (4), 367–380. doi:10.1111/jpi.12176
cardiotoxicity: Effects of preventive strategies. Oxid. Med. Cell. Longev. 2021,
Ho, L., Tan, S. Y., Wee, S., Wu, Y., Tan, S. J., Ramakrishna, N. B., et al. (2015).
8863789. doi:10.1155/2021/8863789
ELABELA is an endogenous growth factor that sustains hESC self-renewal via the
Chen, D., Yu, W., Zhong, C., Hong, Q., Huang, G., Que, D., et al. (2022). Elabela PI3K/AKT pathway. Cell. Stem Cell. 17 (4), 435–447. doi:10.1016/j.stem.2015.
ameliorates doxorubicin-induced cardiotoxicity by promoting autophagic flux through 08.010
TFEB pathway. Pharmacol. Res. 178, 106186. doi:10.1016/j.phrs.2022.106186
Hou, D., Hu, F., Mao, Y., Yan, L., Zhang, Y., Zheng, Z., et al. (2022). Cationic
Chen, H., Wang, L., Wang, W., Cheng, C., Zhang, Y., Zhou, Y., et al. (2017). antimicrobial peptide NRC-03 induces oral squamous cell carcinoma cell apoptosis
ELABELA and an ELABELA fragment protect against AKI. J. Am. Soc. Nephrol. 28 via CypD-mPTP axis-mediated mitochondrial oxidative stress. Redox Biol. 54,
(9), 2694–2707. doi:10.1681/ASN.2016111210 102355. doi:10.1016/j.redox.2022.102355
Chiu, C. F., Ho, M. Y., Peng, J. M., Hung, S. W., Lee, W. H., Liang, C. M., et al. Ishimaru, Y., Shibagaki, F., Yamamuro, A., Yoshioka, Y., and Maeda, S. (2017).
(2013). Raf activation by Ras and promotion of cellular metastasis require An apelin receptor antagonist prevents pathological retinal angiogenesis with
phosphorylation of prohibitin in the raft domain of the plasma membrane. ischemic retinopathy in mice. Sci. Rep. 7 (1), 15062. doi:10.1038/s41598-017-
Oncogene 32 (6), 777–787. doi:10.1038/onc.2012.86 15602-3
Dai, C., Li, J., Tang, S., Li, J., and Xiao, X. (2014). Colistin-induced nephrotoxicity Javadov, S., Choi, A., Rajapurohitam, V., Zeidan, A., Basnakian, A. G., and
in mice involves the mitochondrial, death receptor, and endoplasmic reticulum Karmazyn, M. (2008). NHE-1 inhibition-induced cardioprotection against
ischaemia/reperfusion is associated with attenuation of the mitochondrial Reginauld, S. H., Cannone, V., Iyer, S., Scott, C., Bailey, K., Schaefer, J., et al.
permeability transition. Cardiovasc. Res. 77 (2), 416–424. doi:10.1093/cvr/cvm039 (2019). Differential regulation of ANP and BNP in acute decompensated heart
failure: Deficiency of ANP. JACC. Heart Fail. 7 (10), 891–898. doi:10.1016/j.jchf.
Johnson, T. A., Milan-Lobo, L., Che, T., Ferwerda, M., Lambu, E., McIntosh, N. L.,
2019.05.012
et al. (2017). Identification of the first marine-derived opioid receptor "balanced"
agonist with a signaling profile that resembles the endorphins. ACS Chem. Neurosci. Sato, T., Sato, C., Kadowaki, A., Watanabe, H., Ho, L., Ishida, J., et al. (2017).
8 (3), 473–485. doi:10.1021/acschemneuro.6b00167 ELABELA-APJ axis protects from pressure overload heart failure and angiotensin
II-induced cardiac damage. Cardiovasc. Res. 113 (7), 760–769. doi:10.1093/cvr/
Kostrzewa-Nowak, D., Paine, M. J., Wolf, C. R., and Tarasiuk, J. (2005). The role
cvx061
of bioreductive activation of doxorubicin in cytotoxic activity against leukaemia
HL60-sensitive cell line and its multidrug-resistant sublines. Br. J. Cancer 93 (1), Schieber, M., and Chandel, N. S. (2014). ROS function in redox signaling and
89–97. doi:10.1038/sj.bjc.6602639 oxidative stress. Curr. Biol. 24 (10), R453–R462. doi:10.1016/j.cub.2014.03.034
Lal, H., Ahmad, F., Woodgett, J., and Force, T. (2015). The GSK-3 family as Song, H. P., Chu, Z. G., Zhang, D. X., Dang, Y. M., and Zhang, Q. (2018). PI3K-
therapeutic target for myocardial diseases. Circ. Res. 116 (1), 138–149. doi:10.1161/ AKT pathway protects cardiomyocytes against hypoxia-induced apoptosis by
CIRCRESAHA.116.303613 MitoKATP-mediated mitochondrial translocation of pAKT. Cell. Physiol.
biochem. 49 (2), 717–727. doi:10.1159/000493037
Lee, D., Lee, S. H., Noh, I., Oh, E., Ryu, H., Ha, J., et al. (2019). A helical
polypeptide-based potassium ionophore induces endoplasmic reticulum stress- Sun, X., Guo, C., Zhao, F., Zhu, J., Xu, Y., Liu, Z. Q., et al. (2019). Vasoactive
mediated apoptosis by perturbing ion homeostasis. Adv. Sci. 6 (14), 1801995. intestinal peptide stabilizes intestinal immune homeostasis through maintaining
doi:10.1002/advs.201801995 interleukin-10 expression in regulatory B cells. Theranostics 9 (10), 2800–2811.
doi:10.7150/thno.34414
Leemasawat, K., Phrommintikul, A., Chattipakorn, S. C., and Chattipakorn, N.
(2020). Mechanisms and potential interventions associated with the cardiotoxicity Tocchetti, C. G., Carpi, A., Coppola, C., Quintavalle, C., Rea, D., Campesan, M.,
of ErbB2-targeted drugs: Insights from in vitro, in vivo, and clinical studies in breast et al. (2014). Ranolazine protects from doxorubicin-induced oxidative stress and
cancer patients. Cell. Mol. Life Sci. 77 (8), 1571–1589. doi:10.1007/s00018-019- cardiac dysfunction. Eur. J. Heart Fail. 16 (4), 358–366. doi:10.1002/ejhf.50
03340-w
Wang, Y., Lei, T., Yuan, J., Wu, Y., Shen, X., Gao, J., et al. (2018). GCN2 deficiency
Li, M., Qi, Z., Zhang, J., Zhu, K., and Wang, Y. (2021). Effect and mechanism of ameliorates doxorubicin-induced cardiotoxicity by decreasing cardiomyocyte
Si-Miao-Yong-An on vasa vasorum remodeling in ApoE-/- mice with apoptosis and myocardial oxidative stress. Redox Biol. 17, 25–34. doi:10.1016/j.
atherosclerosis vulnerable plague. Front. Pharmacol. 12, 634611. doi:10.3389/ redox.2018.04.009
fphar.2021.634611
Wu, S. P., Tam, M., Vega, R. M., Perez, C. A., and Gerber, N. K. (2017). Effect of
Lin, Z., Tian, X. Y., Huang, X. X., He, L. L., and Xu, F. (2019). microRNA-186 breast irradiation on cardiac disease in women enrolled in BCIRG-001 at 10-year
inhibition of PI3K-AKT pathway via SPP1 inhibits chondrocyte apoptosis in follow-up. Int. J. Radiat. Oncol. Biol. Phys. 99 (3), 541–548. doi:10.1016/j.ijrobp.
mice with osteoarthritis. J. Cell. Physiol. 234 (5), 6042–6053. doi:10.1002/jcp. 2017.06.018
27225
Xu, C. (2021). The elabela in hypertension, cardiovascular disease, renal disease,
Lüscher, T. F. (2015). Ageing, inflammation, and oxidative stress: Final common and preeclampsia: An update. J. Hypertens. 39 (1), 12–22. doi:10.1097/HJH.
pathways of cardiovascular disease. Eur. Heart J. 36 (48), 3381–3383. doi:10.1093/ 0000000000002591
eurheartj/ehv679
Yang, P., Read, C., Kuc, R. E., Buonincontri, G., Southwood, M., Torella, R., et al.
Ma, Z., Zhao, L., Martin, S., Zhang, Y., Dong, Y., Zhong, J. C., et al. (2021). Lower (2017). Elabela/toddler is an endogenous agonist of the apelin APJ receptor in the
plasma elabela levels in hypertensive patients with heart failure predict the adult cardiovascular system, and exogenous administration of the peptide
occurrence of major adverse cardiac events: A preliminary study. Front. compensates for the downregulation of its expression in pulmonary arterial
Cardiovasc. Med. 8, 638468. doi:10.3389/fcvm.2021.638468 hypertension. Circulation 135 (12), 1160–1173. doi:10.1161/
CIRCULATIONAHA.116.023218
Ma, Z. G., Kong, C. Y., Wu, H. M., Song, P., Zhang, X., Yuan, Y. P., et al.
(2020). Toll-like receptor 5 deficiency diminishes doxorubicin-induced acute Ye, N., Zhang, N., Zhang, Y., Qian, H., Wu, B., and Sun, Y. (2019). Cul4a as a new
cardiotoxicity in mice. Theranostics 10 (24), 11013–11025. doi:10.7150/thno. interaction protein of PARP1 inhibits oxidative stress-induced H9c2 cell apoptosis.
47516 Oxid. Med. Cell. Longev. 2019, 4273261. doi:10.1155/2019/4273261
Malla, J. A., Umesh, R. M., Yousf, S., Mane, S., Sharma, S., Lahiri, M., et al. (2020). Yuan, Y., Fan, S., Shu, L., Huang, W., Xie, L., Bi, C., et al. (2020). Exploration the
A glutathione activatable ion channel induces apoptosis in cancer cells by depleting mechanism of doxorubicin-induced heart failure in rats by integration of
intracellular glutathione levels. Angew. Chem. Int. Ed. Engl. 59 (20), 7944–7952. proteomics and metabolomics data. Front. Pharmacol. 11, 600561. doi:10.3389/
doi:10.1002/anie.202000961 fphar.2020.600561
Münzel, T., Gori, T., Bruno, R. M., and Taddei, S. (2010). Is oxidative stress a Zhang, S., Li, Z. T., Liu, M., Wang, J. R., Xu, M. Q., Li, Z. Y., et al. (2019). Anti-
therapeutic target in cardiovascular disease? Eur. Heart J. 31 (22), 2741–2748. tumour activity of low molecular weight heparin doxorubicin nanoparticles for
doi:10.1093/eurheartj/ehq396 histone H1 high-expressive prostate cancer PC-3M cells. J. Control. Release 295,
102–117. doi:10.1016/j.jconrel.2018.12.034
Paes, W., Leonov, G., Partridge, T., Chikata, T., Murakoshi, H., Frangou, A., et al.
(2019). Contribution of proteasome-catalyzed peptide cis-splicing to viral targeting Zhang, T., Wang, Y., Ma, X., Ouyang, Z., Deng, L., Shen, S., et al. (2022).
by CD8+ T cells in HIV-1 infection. Proc. Natl. Acad. Sci. U. S. A. 116 (49), Melatonin alleviates copper toxicity via improving ROS metabolism and
24748–24759. doi:10.1073/pnas.1911622116 antioxidant defense response in tomato seedlings. Antioxidants (Basel) 11 (4),
758. doi:10.3390/antiox11040758
Read, C., Nyimanu, D., Williams, T. L., Huggins, D. J., Sulentic, P., Macrae, R. G.
C., et al. (2019). International union of basic and clinical Pharmacology. CVII. Zhou, J., Ahmad, F., Parikh, S., Hoffman, N. E., Rajan, S., Verma, V. K., et al.
Structure and Pharmacology of the apelin receptor with a recommendation that (2016). Loss of adult cardiac myocyte GSK-3 leads to mitotic catastrophe resulting
elabela/toddler is a second endogenous peptide ligand. Pharmacol. Rev. 71 (4), in fatal dilated cardiomyopathy. Circ. Res. 118 (8), 1208–1222. doi:10.1161/
467–502. doi:10.1124/pr.119.017533 CIRCRESAHA.116.308544