150
Views
1
CrossRef citations to date
0
Altmetric
ORIGINAL RESEARCH

Evaluation of Ameliorative Effect of Quercetin and Candesartan in Doxorubicin-Induced Cardiotoxicity

, & ORCID Icon
Pages 857-866 | Received 13 Jul 2022, Accepted 05 Dec 2022, Published online: 13 Dec 2022

Abstract

Background

Several mechanisms have been explored for the anthracycline myocardial toxicity. These are free-radical generation, myocyte apoptosis, lipid peroxidation, mitochondrial deterioration, and direct repression of muscle-specific gene expression. Adriamycin (Doxorubicin) is a potent anti-cancer agent. Adriamycin in prolonged use is fatal and generates free radicals that lead to dose-dependent cardiac toxicity.

Objective

The intent of the study was to explore the protective activity of candesartan and quercetin in cardiomyopathy induced by doxorubicin in rats.

Methods

To induce cardiac toxicity, rats were intraperitoneally treated with doxorubicin (06 equivalent injections of 2.5 mg/kg, i. p. at 48 hour interval for 02 consecutive weeks to achieve a cumulative dose of 15 mg/kg). Individual and combined oral treatment of candesartan (5 mg/kg/day) and quercetin (10 mg/kg/day) was administered for four weeks.

Results

Following cardiomyopathy, heart/body weight ratio (3.526 × 10−3), serum creatine kinase (352.4±16.99 IU/L), lactate dehydrogenase (661.7±20.45 IU/L) levels were elevated in addition to altered lipid profile (TC – 118.4±4.25 mg/dL, TG – 263.3±9.99 mg/dL, VLDL – 52.66±1.99 mg/dL, LDL – 52.99±5.80 mg/dL and HDL – 12.78±0.36 mg/dL). The pre-cotreatment of candesartan and quercetin significantly restored the values to normal. The increased level of lipid peroxides (33.12±1.63 µmol/mg protein), serum troponin-T (1.82 ± 0.11 pg/mL) and nitric oxide (13.33±0.73 nmol/mg protein) level along with attenuating antioxidant profile, ie catalase, glutathione and superoxide dismutase (1.43±0.12 nmol/mg protein, 8.48±0.42 nmol/mg protein and 2.09±0.031 U/mg protein) were reversed to normal. Morphometry and histopathologic changes represented a beneficial effect of single and combination pre-cotreatment of drugs which significantly decreases adriamycin cardiac toxicity.

Conclusion

The overall result depicts more beneficial and cardioprotective effect of quercetin and candesartan combination as compared to their individual effects in doxorubicin treated animals. Therefore, this combination might be a suitable option to treat the cardiotoxic effect of doxorubicin.

Introduction

Adriamycin, also termed as Doxorubicin (DOX), is a member of anthracycline antibiotics. It is isolated from Streptomyces peacetius var. Caesius, effectively and extensively used as a broad-spectrum anti-cancer agent. However, the clinical use of DOX is restricted by its acute and chronic cardiotoxicity.Citation1 Various mechanisms have been recommended for anthracyclines myocardial toxicity. It includes free-radical generation, myocyte apoptosis, lipid peroxidation, mitochondrial deterioration and direct repression of muscle-specific gene expression.Citation2,Citation3 Dexrazoxane is a cytoprotective drug used to prevent and improve cardiomyopathy associated with doxorubicin treatment for metastatic breast cancer. But overall usage of DOX must be monitored in various cases.

Oxidative stress triggers the release of cytokines to induce cardiac inflammation or cardiomyopathy.Citation4 It is also linked to rise in cardiac angiotensin converting enzyme (ACE) activity. ACE inhibition drastically declines adriamycin toxicity in clinicalCitation5,Citation6 and pre-clinical studies.Citation7–9 Ang II enhances cytokine production eg tumor necrosis factor-α (TNF- α) via AT1 receptors present on monocytes, macrophages and vascular smooth muscle cells.Citation10 Ang II play a significant role in the pathology of hypertension and IHD (ischemic heart disease). The cardiac remodelling process is significant with a decrease Ang II activity along with long-term survival in pre-clinical and clinical of cardiac hypertrophy and failure.Citation11,Citation12 AT1 also aggravates cardiac disorders like left ventricular (LV) hypertrophy, myocardial infarction, atherosclerosis, hypertension, and heart failure. Candesartan (CAN) an ARB, potentiates AT2 receptor activity and opposes AT1 stimulation.Citation13–15

Nowadays, edibles from medicinal source are gaining popularity due to their protective effects in cardiovascular disease therapy.Citation16 Quercetin (QRN) is widely and commonly ingested dietary polyphenolic flavonoid.Citation17,Citation18 It possesses various pharmacological actions, including antiviral,Citation19 antidiabetic,Citation20 anti-inflammatory,Citation21 cardioprotective,Citation22 neuroprotectiveCitation23 and antiproliferative activity.Citation24 Adequate presence of QRN in food such as fruits, vegetables, wine, etc.Citation25 up surged the interest for cardioprotective activity of natural substances.Citation26 Due to oxidative stress, many reactive oxygen species (ROS) are produced, leading to heart diseases like atherosclerosis, IHD, heart failure and hypertension. QRN has high antioxidant properties among all the flavonoids to prevent oxidative stress.Citation27,Citation28

The current protocol examines the consequences of single and combined pre-cotreatment of QRN and CAN on DOX induced cardiac toxicity. QRN (directly captures ROS to show free radical neutralizing activity) and CAN (blocks RAAS by AT1R and improves ROS induced oxidative stress) could produce additional protective effects against chronic DOX cardiotoxicity. Also, candesartan half-life is 9 hrs, and 10,000-fold AT1 affinity as compared to AT2 whereas quercetin has a half-life is ~12.5 hrs as per studies.Citation29–31 This investigation can show the probable preliminary mechanisms underlying these effects.

Materials and Methods

Doxorubicin hydrochloride vial 50mg was purchased from Aaltra Med Health care limited, Hyderabad, India. Quercetin powder 25 g was purchased from Sigma–Aldrich, St. Louis, MO, USA. Other chemicals were purchased from CDH, Hi Media, Reckon diagnostics and ERBA diagnostics. All chemicals used were of analytical grade.

Experimental Design of in-vivo Study

Adult male Wister rats, weighing 200–250 g were used after a one-week acclimatization period. The experimental protocol for animals was performed as per the guidelines of Committee for the purpose of control and supervision of experiments on animals (CPCSEA). The protocol for in-vivo study was approved by Institutional Animal Ethical Committee (IAEC) of Institute of Technology and Science (ITS), College of Pharmacy, Muradnagar, Ghaziabad, UP, India. The study protocol number was ITS/07/IAEC/2013. During the study period, standard environmental conditions viz.12h dark/light cycle and temperature (21 ± 2°C) were maintained. Animals had free access to standard laboratory pellets and water ad libitum. Rats were divided into five groups (08 rats/group) and were administered with drugs viz. DOX, CAN, and QRN as shown in . Such type of pattern is reported previouslyCitation32,Citation33 in .

Table 1 Route, Dose and Description of Drug Administration

Serum and Tissue Sampling

Rats were sacrificed after 24-hours of last DOX injection by cervical dislocation. Blood was collected and serum was separated by centrifugation at 10,000×g for 10 min. Heart of animals were removed and washed by ice-cold saline. The heart was weighed and homogenized in ice-cold saline to obtain 10% (w/v) homogenate. Serum and heart homogenate were stored at −80°C, until analysis. The heart: body weight ratio was calculated for gravimetric analysis, by weight of the animals before sacrifice and of isolated hearts using a precision balance.

Estimation of Serum Creatine Kinase (CK-MB) and Lactate Dehydrogenase (LDH) Levels

CK-MB found in high concentration in myocardium (14–42%) was determined using diagnostic kit (ERBA diagnostics). Mix and incubate for 3 mins at 37°C. Then, measure the absorbance (340 nm) at time 0 and again exactly after 1, 2 and 3 minutes. The absorbance change (∆A) at 60 second interval was measured using UV spectrometry to calculate CK-MB level as (U/L). LDH, an cytoplasmic enzyme, was determined using diagnostic kit (Reckon diagnostics). Estimation of these enzymes provides a strong base to monitor the progress of onset and status in CHD patients. Mix and read first absorbance of the test exactly at one minute and thereafter at 30, 60 and 90 seconds at 340 nm. Determine the mean change in absorbance per minute and calculate test results as (IU/L).Citation27,Citation28

Estimation of Myocardial Antioxidant Enzyme Activities

Commercial kits were used to estimate myocardial SOD, CAT, GSH activity by colorimetric method. The % inhibition was reported to calculate SOD (U/mg protein), CAT and GSH (nmol/mg protein) concentration.Citation28 Thiobarbituric acid reactive species (TBARS), also termed as malondialdehyde (MDA) is a marker of oxidative stress, can be determined in cardiac homogenates by lipid peroxidation. It is measured by UV spectrometer at 532nm.

Estimation of Lipid Profile

Commercial kits were used to determine serum triglyceride (TG), total cholesterol (TC) and high-density lipoprotein (HDL). Friedewald’s formula was employed to determine low-density lipoprotein (LDL) and very low-density lipoprotein (VLDL).Citation34

Estimation of Myocardial Total Nitrite/Nitrate and Cardiac Troponin T Concentration

Griess reaction was used for the colorimetric assay of nitrate and nitrite (NO indicators in tissue). Total nitrite is then measured as it is proportionately converted into nitrite and nitrate.Citation35 Cardiac troponin T (cTnT) was determined using commercial kit and as per manufacturer’s guidelines.

Morphometric Analysis

Cardiac tissues were taken in thin slices and stained by 1% TTC (triphenyl tetrazolium chloride) in phosphate buffer saline (pH 7.4).Citation36 Viable tissues appear red due to staining whereas no stains were visible on non-viable tissue. The stained heart images were analysed for area of necrosis.

Histopathological Examination of the Cardiac Tissues

Twenty-four hours after last DOX injection, the rats were sacrificed and hearts were isolated. Immediately after isolation, the heart tissue was washed with cold phosphate buffer saline (pH = 7.4) and fixed in 10% buffered formalin. Further, haematoxylin and eosin stain were used and 5μm stained sections were observed under a light microscope. Histopathological changes were identified through the images taken by digital camera.Citation36

Statistical Analysis

Results were expressed as mean ± standard error of the mean (S.E.M). Statistical significance was determined by one-way analysis of variance (ANOVA) followed by Tukey–Kramer post-test to compare all groups. Results were considered to be statistically significant when P < 0.001, 0.01 and 0.05.

Results

General Data (Heart/Body Weight Ratio)

The results indicated a significant rise (P < 0.05) in ratio in DOX-treated group (3.526 × 10−3) as compared to control (2.145 × 10−3) in . QRN administration (2.767 × 10−3) significantly reduced the heart/body weight ratio (P < 0.05), while combination of CAN and QRN (2.276 × 10−3) resulted in a more pronounced decline in heart/body weight ratio at P < 0.05.

Table 2 Effect of CAN, QRN and CAND+ QRN on Heart/Body Weight Ratio

Effect of the Single and Combined Pre-Cotreatment of CAN and QRN on the Serum CK-MB and LDH Levels

DOX caused a significant rise (P < 0.01) in serum CK-MB and LDH levels as 352.4±16.99 IU/L and 661.7±20.45 IU/L, respectively, as shown in . CAN or QRN individually suggested a significant decrease in serum CK-MB and LDH levels as compared to DOX-treated rats. Further, pre-cotreatment showed enhanced effect 164.7±7.605 IU/L and 251.3±20.49 IU/L rather than the individuals.

Table 3 Effect of CAN, QRN and CAND+ QRN on Plasma Markers of Cardiac Damage

Effect of Individual and Combined Pre-Cotreatment of CAN and QRN on Myocardial Antioxidant Levels, Lipid Peroxidation and Total Nitrite Levels

DOX treated animals showed a marked decrease in myocardial antioxidant activity ie CAT, GSH and SOD (1.43±0.12 nmol/mg protein, 8.48±0.42 nmol/mg protein and 2.09±0.031 U/mg protein) at p<0.001 as compared to control (4.08±0.12 nmol/mg protein, 28.29±0.43 nmol/mg protein and 5.51±0.088 U/mg protein) in . CAN or QRN single administration improved CAT, GSH and SOD levels. This effect was markedly enhanced viz. 3.54±0.32 nmol/mg protein, 23.12±0.64 nmol/mg protein and 3.23±0.015 U/mg protein on collateral administration (p<0.001).

Table 4 Effect of CAN, QRN and CAND+ QRN on Antioxidant Level

DOX induced a significant rise (P < 0.01) in myocardial MDA, 33.12±1.63 µmol/mg protein and NO, 13.33±0.73 nmol/mg protein level as compared to control 19.17±1.20 µmol/mg protein and 5.62±0.61 nmol/mg protein, respectively, . The combined pre-cotreatment of CAN and QRN resulted in a more significant (P < 0.01) decrease in MDA and NO levels ie 18.59±0.65 µmol/mg protein and 5.80±0.39 nmol/mg protein.

Table 5 Effect of CAN, QRN and CAN+ QRN on Cardiac LPO Levels and NO Level

Effects of Individual and Combined Pre-Cotreatment of CAN and QRN on Lipoprotein Fractions and Serum Troponin-T Levels

DOX induction resulted in a significant rise (p<0.001) in the levels of TC, TG, VLDL and LDL (118.4±4.25 mg/dL, 263.3±9.99 mg/dL, 52.66±1.99 mg/dL and 52.99±5.80 mg/dL) along with significant decrease in HDL cholesterol levels (12.78±0.36 mg/dL), and . Pre-treatment with CAN and QRN individually significantly (p<0.001) lowered TC, TG, LDL, VLDL and increased HDL level in serum. Pre-cotreatment of combination significantly (p<0.001) lowered TC, TG, VLDL, LDL and increased HDL level in serum, ie 81.83±3.59 mg/dL, 109.2±8.97 mg/dL, 21.84±1.79 mg/dL, 21.29±3.96 mg/dL and 19.08±2.20 mg/dL, respectively.

Table 6 Effect of CAN, QRN and CAN+QRN on Plasma Lipid Profile

Table 7 Effect of CAN, QRN and CAN+QRN on Plasma Lipid Profile and Serum Troponin-T Levels

Rats treated with DOX showed increased serum cTnT level, 1.82 ± 0.11 pg/mL suggesting myocardial lesions (p<0.001), . Groups treated with CAN (0.39 ± 0.03 pg/mL) or QRN (0.42 ± 0.01 pg/mL) or the combination (0.34 ± 0.02 pg/mL) showed significant reduction (p<0.01) in serum cTnT level compared to the DOX-treated group.

Morphometry and Histopathological Analysis

TTC staining of heart of control rats showed brick red coloration indicative of a greater number of viable cells. Rats treated with doxorubicin exhibited pale coloration indicating areas of necrosis as shown in . However, Doxorubicin rats pre-treated with candesartan, quercetin and the combination showed a protective effect with a minimal pale coloration. Histological analysis of control group showed normal cardiac tissue without any myonecrosis. DOX-treated group developed cardiac necrosis with oedema, leukocyte infiltration and intramuscular haemorrhage. The single and pre-cotreatment of CAN and QRN ameliorated the histopathological changes of cardiomyocytes in .

Figure 1 Effect of CAN, QRN, and CAN+ QRN on morphometry of cardiac tissue. Yellow arrows depict brick red coloration indicative of a greater number of viable cells in treatment groups.

Abbreviations: DOX, doxorubicin; QRN, quercetin; CAN, candesartan.
Figure 1 Effect of CAN, QRN, and CAN+ QRN on morphometry of cardiac tissue. Yellow arrows depict brick red coloration indicative of a greater number of viable cells in treatment groups.

Figure 2 Effect of CAN, QRN, and CAN+ QRN on histopathology of cardiac tissue. (DOX – doxorubicin; QRN – quercetin; CAN – candesartan) Blue arrow depicts more amount of leukocyte infiltration and intramuscular hemorrhage in DOX treated group as compared to the treatment groups.

Figure 2 Effect of CAN, QRN, and CAN+ QRN on histopathology of cardiac tissue. (DOX – doxorubicin; QRN – quercetin; CAN – candesartan) Blue arrow depicts more amount of leukocyte infiltration and intramuscular hemorrhage in DOX treated group as compared to the treatment groups.

Discussion

Doxorubicin (DOX) is one of the main anticancer drugs that can induce oxidative damage to cardiac tissues. It mainly targets the cardiac antioxidant defence mechanism. In recent times, many endogenous myocardial antioxidants have become promising therapeutic targets of treatment associated with increased oxidative stress.Citation37 Doxorubicin causes rats to lose weight over a period following injection, as confirmed by other reports.Citation38 DOX-induced cardiac toxicity is identified by decreased body weight and increased heart weight.Citation39 This study confirms the earlier findings.

One of the important markers of early and late-stage cardiac injury is CK-MB (specific for cardiac muscle)Citation40. In the study, a reasonable increase in serum cardiac isoenzymes (LDH and CK-MB) was observed due to DOX-induced cardiac necrosis. CAN inhibits AT1R-mediated ROS generation, thereby preventing CK-MB and LDH leakage.Citation41 QRN inhibit angiotensin converting enzyme activity, thereby decreasing the conversion of Ang I to Ang II.Citation42 The results indicate that QRN pre-treatment can potentiate the cardioprotective effect of CAN due to its free radical scavenging and antioxidant properties. QRN and its derivatives may represent promising cardioprotective activity for prevention and treatment of wide range of cardiac disease.Citation43

Rise in lipid peroxidation products is considered as biochemical marker of oxidative stress in DOX-treated rats. Due to lipid peroxidation, inflammatory cells accumulate in cardiac myocytes.Citation44 Thus, inactivity of GSH, SOD and CAT in DOX-treated group is due to superoxide anion generation at the site of damage in myocardium. Current study showed that combined effect of CAN and QRN treatment could decrease the lipid peroxide levelsCitation33 by neutralizing ROS.

CAN treatment could decline DOX-induced elevation of myocardial NO levels. This could be due to decreased TNF-α level and blockade of Ang II-mediated elevation of ROS. Our data showed that CAN produced more pronounced effect in lowering NO levels than QRN. The probable reason for this effect can be that AT1R blockade increases circulating levels of Ang II that could in theory act on the unopposed AT2R effects.Citation45

Lipids consist of cholesterol (HDL and LDL cholesterol), triglycerides, phospholipids and free fatty acids. Observations concluded a significant rise of serum total cholesterol, triglyceride LDL and VLDL in our studies. This showed that DOX reduced the rate of lipolysis, which is deleterious for heart functionCitation46,Citation47 Any change in lipid metabolism directly alters the concentration of lipoproteins. The present combination showed a potent antioxidant activity. It stabilizes lipid metabolizing enzymes by its hypolipidemic effect thereby proving its potential as a cardio-protector.

Metabolically viable cells and tissues can be easily identified using TTC staining (redox indicator). It provides the infarct size to assess myonecrosis of cardiac tissue. The myocardial respiration generates enzymes that reduces TTC to a brick red precipitate ie TPF (1,3,5-triphenylformazan). Thus, viable tissues appear red as compared to the pale non-viable tissues.Citation48 The single or combined pre-cotreatment of CAN and QRN portrayed minimal pale-coloration demonstrating normal cardiac tissues. Histoarchitectural observations verifies DOX-induced necrosis in cardiac fibers. These detrimental changes are remarkably countered by pre-treatment with CAN, QRN or both providing maximal safeguard. Myocardial lesions were diminished to marked extent in groups treated with CAN, QRN or both providing microscopic evidences of cardioprotective.Citation31,Citation43

Conclusion

In conclusion, our study provides an explanation for the cardioprotective effect of quercetin individually as well as in combination with candesartan in doxorubicin-treated animals. Quercetin’s antioxidant properties might have shown the additional effect to candesartan. The results indicate that QRN pre-treatment can potentiate the cardioprotective effect of CAN due to its free radical scavenging and antioxidant properties. Therefore, this combination might be a suitable option to treat the cardiotoxic effect of doxorubicin. Nevertheless, in future additional studies are required to demonstrate and approve the efficacy and safety of quercetin/candesartan combination regimens in clinical practice.

Abbreviations

CAN, candesartan; QRN, quercetin; DOX, doxorubicin; CK-MB, serum creatine kinase; LDH, lactate dehydrogenase; ACE, angiotensin-converting enzyme; AT, angiotensin II receptor; GSH, reduced glutathione; MDA, malondialdehyde; SOD, superoxide dismutase; CAT, catalase; ROS, reactive oxygen species; cTnT, serum troponin-T; NO, nitric oxide.

Human and Animal Rights

No humans were involved in the study. All animal-related experiments were conducted as per the guidelines by CPCSEA (Committee for the purpose of control and supervision of experiments on animals). The study protocol (ITS/07/IAEC/2013) was approved by members of the Institutional Animal Ethical Committee (IAEC).

Ethics Approval and Consent to Participate

The experimental protocol for animals was performed as per the guidelines of Committee for the purpose of control and supervision of experiments on animals (CPCSEA). The protocol for in-vivo study was approved by Institutional Animal Ethical Committee (IAEC) of Institute of Technology and Science (ITS), College of Pharmacy, Muradnagar, Ghaziabad, UP, India. The study protocol number was ITS/07/IAEC/2013.

Disclosure

The authors declare that there are no conflicts of interest regarding the publication of this paper.

Acknowledgment

The authors are grateful to the management of I.T.S College of Pharmacy for providing the necessary support for the successful completion of the work.

Additional information

Funding

The project was non-funded and self-financed.

References

  • Abu-Gazia M, El-Magd MA. Ameliorative effect of cardamom aqueous extract on doxorubicin-induced cardiotoxicity in rats. Cells Tissues Organs. 2018;206:62–72. doi:10.1159/000496109
  • Sawyer DB, Fukazawa R, Arstall MA, Kelly RA. Daunorubicin induced apoptosis in rat cardiac myocytes is inhibited by dexrazoxane. Circ Res. 1999;84:257–265. doi:10.1161/01.RES.84.3.257
  • Hashish FE, Abdel-Wahed MM, El-Odemi MH, El-Naidany SS, ElBatsh MM. Possible protective effects of quercetin on doxorubicin-induced cardiotoxicity in rats. Menoufia Med J. 2021;34:333–339. doi:10.4103/mmj.mmj_5_20
  • Bien S, Riad A, Ritter CA, et al. The endothelin receptor blocker bosentan inhibits doxorubicin-induced cardiomyopathy. Cancer Res. 2007;67:10428–10435. doi:10.1158/0008-5472.CAN-07-1344
  • Jensen BV, Nielsen SL, Skovsgaard T. Treatment with angiotensin converting- enzyme inhibitor for epirubicin-induced dilated cardiomyopathy. Lancet. 1996;347(8997):297–299. doi:10.1016/S0140-6736(96)90469-9
  • Jensen BV, Nielsen SL, Jensen TS. Angiotensin-converting enzyme inhibitor in the treatment of epirubicin-induced dilated cardiomyopathy. Ugeskr Laeger. 1997;159(13):1945–1949.
  • Sacco G, Bigioni M, Evangelista S, Goso C, Manzini S, Maggi CA. Cardioprotective effects of zofenopril, a new angiotensin-converting enzyme inhibitor, on doxorubicin-induced cardiotoxicity in the rat. Eur J Pharmacol. 2001;414(1):71–78. doi:10.1016/S0014-2999(01)00782-8
  • Hauser M, Wilson N. Anthracycline induced cardiomyopathy: successful treatment with angiotensin converting enzyme inhibitors. Eur J Pediatr. 2000;159(5):389. doi:10.1007/s004310051294
  • Hatake K, Miura Y. Angiotensin-converting enzyme inhibitor for epirubicin-induced dilated cardiomyopathy. Lancet. 1996;347(9013):1485. doi:10.1016/S0140-6736(96)91721-3
  • Hahn AW, Jonas U, Buhler FR, Resink TJ. Activation of human peripheral monocytes by angiotensin II. FEBS Lett. 1994;347:178–180. doi:10.1016/0014-5793(94)00531-1
  • Kawabata H, Ryomoto T, Ishikawa K. Cardioprotection with angiotensin converting enzyme inhibitor and angiotensin II type 1 receptor antagonist is not abolished by nitric oxide synthase inhibitor in ischemia-reperfused rabbit hearts. Hypertens Res. 2001;24:403–409. doi:10.1291/hypres.24.403
  • Wang P, Li HW, Wang YP, Chen H, Zhang P. Effects of recombinant human relaxin upon proliferation of cardiac fibroblast and synthesis of collagen under high glucose condition. J Endocrinol Invest. 2009;32:242–247. doi:10.1007/BF03346460
  • Burnier M. Angiotensin II type 1 receptor blockers. Circ J. 2001;103:904–912. doi:10.1161/01.CIR.103.6.904
  • Hoogwerf BJ. Renin–angiotensin system blockade and cardiovascular and renal protection. Am J Cardiol. 2010;105:30A–35A. doi:10.1016/j.amjcard.2009.10.009
  • Venkatesh RC, Ravindra B, Siva RC, Benito J, Maheswari C. Pharmacokinetic interaction study between quercetin and valsartan in rats and in vitro models. Drug Dev Ind Pharm. 2013;39(6):865–872. doi:10.3109/03639045.2012.693502
  • Syahputra RA, Harahap U, Dalimunthe A, Nasution MP, Satria D. The role of flavonoids as a cardioprotective strategy against doxorubicin-induced cardiotoxicity: a review. Molecules. 2022;27(4):1320. doi:10.3390/molecules27041320
  • Tene K, Kalyan Kumar M, Basveshwar G, et al. Polyphenolic rich compounds from Dillenia pentagyna (Roxb) attenuates the doxorubicin-induced cardiotoxicity: a high-frequency ultrasonography assisted approach. Front Pharmacol. 2021;12:624706. doi:10.3389/fphar.2021.624706
  • Boots AW, Haenen GRMM, Bast A. Health effects of quercetin: from antioxidant to nutraceutical. Eur J Pharmacol. 2008;585:325–337. doi:10.1016/j.ejphar.2008.03.008
  • Ohnishi E, Bannai H. Quercetin potentiates TNF-induced antiviral activity. Antiviral Res. 1993;22:327–331. doi:10.1016/0166-3542(93)90041-G
  • Vessal M, Hemmati M, Vasei M. Antidiabetic effects of quercetin in stz-induced diabetic rats. Comp. Biochem. Physiol. C Toxicol Pharmacol. 2003;135:357–364.
  • Rotelli AE, Guardia T, Juarez AO, DelaRocha NE, Pelzer LE. Comparative study of flavonoids in experimental models of inflammation. Pharmacol Res. 2003;48:601–606. doi:10.1016/S1043-6618(03)00225-1
  • Alasmari AF. Cardioprotective and nephroprotective effects of Quercetin against different toxic agents. Eur Rev Med Pharmacol Sci. 2021;25:7425–7439. doi:10.26355/eurrev_202112_27440
  • Dok-Go H, Lee KH, Kim HJ, et al. Neuroprotective effects of antioxidative flavonoids, quercetin, (+)-dihydroquercetinandquercetin3-methylether, isolated from Opuntia ficus-indica var. saboten. Brain Res. 2003;965:130–136. doi:10.1016/S0006-8993(02)04150-1
  • Garcia MV, Crespo I, Collado PS, et al. The anti-inflammatory flavones quercetin and kaempferol cause inhibition of inducible nitric oxide synthase, cyclooxygenase-2 and reactive C-protein, and down-regulation of the nuclear factor kappa B pathway in Chang liver cells. Eur J Pharmacol. 2007;557:221–229. doi:10.1016/j.ejphar.2006.11.014
  • Sharma A, Parikh M, Shah H, Gandhi T. Modulation of Nrf2 by quercetin in doxorubicin-treated rats. Heliyon. 2020;6(4):e03803. doi:10.1016/j.heliyon.2020.e03803
  • Hertog MG, Bueno-de-mesquita HB, Fehily AM, Sweetnam PM, Elwood PC, Kromhout D. Fruit and vegetable consumption and cancer mortality in the Caerphilly Study. Cancer Epidemiol Biomarkers Prev. 1996;5:673–677.
  • Mariee AD, Abd-Allah GM, El-Beshbishy HA. Protective effect of dietary flavonoid quercetin against lipemic–oxidative hepatic injury in hypercholesterolemic rats. Pharm Biol. 2012;50:1019–1025. doi:10.3109/13880209.2012.655424
  • Larson A, Witman MAH, Guo Y, et al. Acute, quercetin-induced reductions in blood pressure in hypertensive individuals are not secondary to lower plasma angiotensin- converting enzyme activity or endothelin-1: nitric oxide. Nutr Res. 2012;32:557–564. doi:10.1016/j.nutres.2012.06.018
  • Munger MA. Use of angiotensin receptor blockers in cardiovascular protection. Current evidence and future directions. Cardiovascul TherPrevent. 2011;10(7):93–104.
  • Moon YJ, Wanga L, DiCenzob R, Morris ME. Quercetin pharmacokinetics in humans. Biopharm Drug Dispos. 2008;4:205–217. doi:10.1002/bdd.605
  • Sobczuk P, Czerwinska M, Kleibert M, Agnieszka C. Anthracycline-induced cardiotoxicity and renin-angiotensin-aldosterone system—from molecular mechanisms to therapeutic applications. Heart Fail Rev. 2022;27:295–319. doi:10.1007/s10741-020-09977-1
  • Haleagrahara N, Radhakrishnan A, Lee N, Kumar P. Flavonoid quercetin protects against swimming stress-induced changes in oxidative biomarkers in the hypothalamus of rats. Eur J Pharmacol. 2009;621:46–52. doi:10.1016/j.ejphar.2009.08.030
  • Soga M, Kamal F, Watanabe K, et al. Effects of angiotensin II receptor blocker (candesartan) in daunorubicin-induced cardiomyopathic rats. Int J Cardiol. 2006;110(3):378–385. doi:10.1016/j.ijcard.2005.08.061
  • Friedewald WT, Levy RI, Fredrickson DS. Estimation of the concentration of low- density lipoprotein cholesterol in plasma, without use of the preparative ultracentrifuge. Clin Chem. 1972;18:499–502. doi:10.1093/clinchem/18.6.499
  • Tsikas D. Analysis of nitrite and nitrate in biological fluids by assays based on the Griess reaction: appraisal of the Griess reaction in the l-arginine/nitric oxide area of research. J Chromatogr B. 2007;851:51–70. doi:10.1016/j.jchromb.2006.07.054
  • Ytrehus K, Liu Y, Tsuchida A, et al. Rat and rabbit heart infarction: effects of anaesthesia, perfusate, risk zone, and method of infarct sizing. Am J of Physiol. 1994;267:H2383–H2390.
  • Elblehi SS, El-Sayed YS, Soliman MM, Shukry M. Date palm pollen extract avert doxorubicin induced cardiomyopathy fibrosis and associated oxidative/nitrosative stress, inflammatory cascade, and apoptosis-targeting bax/bcl-2 and caspase-3 signaling pathways. Animals. 2021;11:88–92. doi:10.3390/ani11030886
  • Rephaeli A, Waks-Yona S, Nudelman A, et al. Anticancer prodrugs of butyric acid and formaldehyde protect against doxorubicin-induced cardiotoxicity. Br J Cancer. 2007;96:1667–1674. doi:10.1038/sj.bjc.6603781
  • Antonio A, Jose M, Jose S, et al. Endurance training attenuates doxorubicin-induced cardiac oxidative damage in mice. Int J Cardiol. 2005;100:451–460. doi:10.1016/j.ijcard.2004.11.004
  • Fadillioglu E, Erdogan H. Effects of erdosteine treatment against doxorubicin-induced toxicity through erythrocyte and plasma oxidant/antioxidant status in rats. Pharmacol Res. 2003;47:317–322. doi:10.1016/S1043-6618(03)00010-0
  • Griendling KK, Minieri CA, Ollerenshaw JD, Alexander RW. Angiotensin II stimulates NADH and NADPH oxidase activity in cultured vascular smooth muscle cells. Circ Res. 1994;74:1141–1148. doi:10.1161/01.RES.74.6.1141
  • Loizzo MR, Said A, Tundis R, et al. Inhibition of angiotensin converting enzyme (ACE) by flavonoids isolated from Ailanthus excelsa (Roxb) (Simaroubaceae). Phytother Res. 2007;21:32–36. doi:10.1002/ptr.2008
  • Ferenczyova K, Kalocayova B, Bartekova M. Potential Implications of Quercetin and its Derivatives in Cardioprotection. Int J Mol Sci. 2020;21(5):1585. doi:10.3390/ijms21051585
  • Saad SY, Najjar TA, Al-Rikabi AC. The preventive role of deferoxamine against acute doxorubicin-induced cardiac, renal and hepatic toxicity in rats. Pharmacol Res. 2001;43:211–218. doi:10.1006/phrs.2000.0769
  • Widdop RE, Jones ES, Hannan RE, Gaspari TA. Angiotensin AT2 receptors: cardiovascular hope or hype? Br J Pharmacol. 2003;140:809–824. doi:10.1038/sj.bjp.0705448
  • Iliskovic SPK. Lipid lowering: an important factor in preventing doxorubicin-induced heart failure. Am J Pathol. 1997;150:727–734.
  • Koutinos G, Stathopoulos GP, Dontas I, et al. The effect of doxorubicin and its analogue mitoxantrone on cardiac muscle and on serum lipids: an experimental study. Anticancer Res. 2002;22:815–820.
  • Altman FP. Tetrazolium salts and formazans. Prog Histochem Cytochem. 1976;9:1–56. doi:10.1016/S0079-6336(76)80015-0