Next Article in Journal
Synthesis, Characterization, and Catalytic Exploration of Mononuclear Mo(VI) Dioxido Complexes of (Z)-1-R-2-(4′,4′-Dimethyl-2′-oxazolin-2′-yl)-eth-1-en-1-ates
Previous Article in Journal
Protective Effect of Quercetin 3-O-Glucuronide against Cisplatin Cytotoxicity in Renal Tubular Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Flavonoids as a Cardioprotective Strategy against Doxorubicin-Induced Cardiotoxicity: A Review

by
Rony Abdi Syahputra
1,*,
Urip Harahap
1,*,
Aminah Dalimunthe
1,
M. Pandapotan Nasution
2 and
Denny Satria
2
1
Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan 20155, Indonesia
2
Department of Pharmaceutical Biology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan 20155, Indonesia
*
Authors to whom correspondence should be addressed.
Molecules 2022, 27(4), 1320; https://doi.org/10.3390/molecules27041320
Submission received: 17 January 2022 / Revised: 26 January 2022 / Accepted: 2 February 2022 / Published: 15 February 2022

Abstract

:
Doxorubicin is a widely used and promising anticancer drug; however, a severe dose-dependent cardiotoxicity hampers its therapeutic value. Doxorubicin may cause acute and chronic issues, depending on the duration of toxicity. In clinical practice, the accumulative toxic dose is up to 400 mg/m2 and increasing the dose will increase the probability of cardiac toxicity. Several molecular mechanisms underlying the pathogenesis of doxorubicin cardiotoxicity have been proposed, including oxidative stress, topoisomerase beta II inhibition, mitochondrial dysfunction, Ca2+ homeostasis dysregulation, intracellular iron accumulation, ensuing cell death (apoptosis and necrosis), autophagy, and myofibrillar disarray and loss. Natural products including flavonoids have been widely studied both in cell, animal, and human models which proves that flavonoids alleviate cardiac toxicity caused by doxorubicin. This review comprehensively summarizes cardioprotective activity flavonoids including quercetin, luteolin, rutin, apigenin, naringenin, and hesperidin against doxorubicin, both in in vitro and in vivo models.

1. Introduction

Doxorubicin is a part of the anthracycline group of chemotherapy, one of the most widely used and efficacious methods for treating hematological malignancies, solid tumors, and lymphoma [1]. The essential mechanism of doxorubicin involves generating oxidator and inhibiting topoisomerase II in cancer cells, although, on the other hand, it is toxic to several organs, including the heart [2]. Therefore, a severe dose-dependent cardiotoxicity hampers its therapeutic value. Based on the administration duration of doxorubicin, its cardiotoxicity is divided into acute and chronic toxicity, with acute toxicity occurring during the early administration of doxorubicin (within 2–3 days), in approximately 11% of incidences. Clinical manifestations of acute toxicity are hypotension, tachycardia and various arrhythmias, pericarditis, or myocarditis, but these are reversible with appropriate treatment [3,4]. Chronic toxicity occurs after 30 days of the administration of doxorubicin, but the percentage of toxicity is lesser than acute toxicity—about 1.7%. This can lead to left ventricular dysfunction that irreversibly evolves toward congestive heart failure [5].
Cardiotoxicity due to doxorubicin depends on the dose administration: normally a dose below 400 mg/m2 is less toxic. If the dose exceeds more than 400 mg/m2, it will increase the likelihood of toxicity occurrence [6]. In addition, medical history and age also influence the risk of cardiotoxicity. A history of diabetic or cardiovascular disease such as hypertension, hyperlipidemia, or atherosclerosis develops these complications [7]. Moreover, age >65 and >4 years old are the most vulnerable [8]. Tobacco use, poor nutrition, or being physically inactive also have a part in developing the risk of cardiotoxicity [9].
Several molecular mechanisms underlying the pathogenesis of doxorubicin cardiotoxicity have been proposed, including oxidative stress [10], topoisomerase II inhibition [11], mitochondrial dysfunction [12], Ca2+ homeostasis dysregulation [13], intracellular iron accumulation [14], ensuing cell death (apoptosis and necrosis) [15], autophagy [16], and myofibrillar disarray and loss [17].
Nowadays, preventive strategies have been delivered to reduce and prevent cardiotoxicity development. These are limited doxorubicin dose [18], liposomal formulation doxorubicin [19], and the co-administration of dexrazoxane with one of the cardioprotective FDA-approved drugs for preventing doxorubicin-induced cardiotoxicity. Up-to-date mechanisms of dexrazoxane are iron chelator, diminishing oxidative stress, and directly competing with topoisomerase II in the nucleus [20]. The early detection of cardiac biomarkers will be beneficial for patients. Cardiac biomarkers use for analysis such as cardiac troponin T (cTnT), brain natriuretic peptide (BNP), atrial natriuretic peptide (ANP), and c-reactive protein (CRP) which are known increase in cardiotoxic patients [21].
Natural products have traditionally been used by humans for food consumption, nutrition, and preventive measures and treatments for several diseases [22]. Flavonoid derived-plants mainly explored and investigated for cardioprotective measures against doxorubicin-induced cardiotoxicity [23]. Flavonoids are divided into the following classes: flavones, flavonols, flavanones, flavanonols, flavanols or catechins, anthocyanins, and chalcones. Each subclass of flavonids has been widely tested on in vitro and in vivo models of cardiotoxicity-induced doxorubicin [24]. Flavonoid has pharmacological effects such as antioxidant, anti-inflammatory, anti-apoptosis, anti-calcium overload, and iron scavenging properties [25]. The flavonoid divided into subclasses quercetin, rutin, apigenin, and luteolin have been researched the most in in vivo and in vitro models against doxorubicin. Particularly, the mechanism of protective activity is clear and well known; subsequently, increases in the expression of nrf-2, SOD, GSH, and HO-1 inhibit the expression of pro-apoptosis protein cytochrome c, Bax, caspase 3, caspase 7, caspase 9, and inhibit the expression of pro-inflammatory protein TNF, IL-1, IL-6, and Nf-kB [26,27,28,29,30]. Therefore, this comprehensive review provides a summary of the cardioprotective mechanism of flavonoids against cardiotoxicity induced by doxorubicin.

2. Doxorubicin Mechanism of Toxicity

2.1. Doxorubicin Generates Reactive Oxygen Species (ROS)

Doxorubicin is a well-known potent generator of ROS both in cytosolic and mitochondria [31]. The metabolite of doxorubicin is doxorubicin-semiquinone, which rapidly oxidizes oxygen (O2) converted into superoxide anion radical (O2). Unfortunately, this O2 is highly reactive with NO, which can produce peroxynitrite (ONOO). The accumulation of ROS is normally cleared by endogenous antioxidants such as SOD, or exogenous antioxidants such as flavonoids that produce H2O2; however, the existence of iron (Fe2+) can directly convert H2O2 into hydroxyl radical (OH), which is a known Fenton reaction [32,33,34,35]. Moreover, in the case of high cumulative doses of doxorubicin, ROS production is also extremely high, which might cause the degradation of lipids in the membrane, decrease the ATP, induce the opening of MPTP, and sensitize the ryanodine receptor that induces the excessive release of calcium into the cytosol. All these ROS effects lead to the apoptosis of cardiomyocytes [36,37,38]. The supplementation of antioxidants will beneficially reduce the ROS overproduction of doxorubicin. Most of the reviewed research confirmed that flavonoids reduce the production of ROS, both in in vivo and in vitro models, but the mechanism remains unclear and needs to be elucidated [39]

2.2. Mitochondria Injury

Mitochondria is the source of energy for cardiomyocytes, uniquely, with mitochondria levels 60% higher in cardiomyocytes than in the rest of the body’s cells [40]. Hence, the volume of cardiomyocytes consists of 30–40% mitochondria [41]. As mentioned earlier, ROS production in the mitochondria is assumed to be the cause of mitochondria injury [42,43,44]. Doxorubicin has a strong binding affinity to cardiolipin in the inner membrane of mitochondria, that may directly cause the disturbance of the electron transport chain (ETC), which cause increased the ROS dan RNS [45,46,47]. Hence, the pro-oxidant causes opening pore mitochondria resulted release of cytochrome c that initiates apoptosis by activating caspase 3. Furthermore, this mechanism also causes mitochondria swelling, which leads to necrosis and rupture of the outer membrane of mitochondria. Both apoptosis and necrosis primarily cause the death of cardiomyocytes mostly mediated by ROS [48,49,50].

2.3. Topoisomerase 2β (TOP2β)

The main mechanism of doxorubicin as an anticancer agent is the binding to topoisomerase in the nucleus of cancer cells [51,52,53]. Topoisomerase has two isoforms which include 2α (TOP2α), and 2β (TOP2β) topoisomerase. In the tumor cell, doxorubicin binds into TOP2α, resulting in DNA degradation and cell death. Unfortunately, the toxicity of doxorubicin also binds to TOP2β, which is dominantly expressed in the adult cardiomyocytes [51]. These complex doxorubicin-DNA-TOP2β cause double-strand DNA breakage, which causes apoptosis [54,55,56]. Interestingly, doxorubicin seems more susceptible to binding to TOP2β in cancer patients. The accumulation of doxorubicin in cancer patients increases the doxorubicin-induced cardiotoxicity. Primarily, the maintenance of doxorubicin dosage in patients is the most important measure in preventing cardiotoxicity. In relation to the nucleus stress due to doxorubicin, the translocation of erythroid-2-related factor (Nrf2) to the nucleus will be provided by the Keap1-Nrf2 complex, which produces OH-1 as a protector [57,58,59]. Meanwhile, the stress condition activates c-Jun N-terminal kinases (JNKs) and p38-MAPKs by cellular oxidative stress, that also correlate with cardiac pathophysiology and apoptotic cell death [60,61,62].

2.4. Calcium Homeostasis Dysregulation

Regulating the level of Ca2+ levels in the cell are essential for maintaining calcium homeostasis. However, doxorubicin increases the level of intracellular Ca2+ [63,64], and it downregulates the expression level of SERCA2a, leading to a decrease in Ca2+ uptake [65,66,67]. SERCA2a plays a vital role in restoring the excessive amount of Ca2+ in the cytosol, the uncontrolled level of Ca2+ in cells causes the impairment of contractile cardiac muscles [68]. Furthermore, doxorubicin also inhibits the Na+/Ca2+ exchanger [69], and the increase in doxorubicin levels also causes an increase in the expression of the ryanodine receptor channels, which leads to the massive release of Ca2+ [70,71]. Interestingly, SERCA is inhibited by the ROS-mediated S-oxidation of the conserved Cys 674 along with increasing RyR2 [72]. The mitochondria absorb a large portion of the released calcium, and the receptor is known to be sensitive to oxidation due to presence of many thiols [73]. Overloading the mitochondria with calcium can result in mitochondrial malfunction and the induction of a cascade of pro-apoptotic events [74]. Furthermore, doxorubicin causes Ca2+-dependent Ca2+/calmodulin-dependent protein kinase II (CaMKII) activation as the result of promoting apoptosis [75].

2.5. Cardiac Biomarkers Injury

Early detection of cardiac injury is beneficial to the patient in preventing the incidence of cardiotoxicity by doxorubicin. Many cardiac biomarkers have been used to predict a cardiotoxic event, one of which includes AST as the first biomarker to assess the cardiac injury. However, the limitation of this is that the release of AST occurs not only in the cardiac cell, but also in liver injury [76,77,78]. Moreover, LDH has been used as a biomarker for cardiac enzymes in the past; usually, the increase in the level of LDH is detected after 24–72 h of cardiac injuries [79]. Meanwhile, in terms of acute myocardial injury, CK-MB is more sensitive compared to LDH, and it is becoming an essential cardiac biomarker of injury [80]. CK-MB levels are higher in cardiac muscle rather than in skeletal muscle, which is primarily made up of CK-MM [81]. Presently, AST, LDH, and CK-MB are no longer recommended as early detectors of cardiac injury, but Troponin is used as an early detector (gold standard) instead [82]. Furthermore, Troponin is divided into three isoforms, namely the troponin T complex of the actin filament, the troponin c complex of Ca binding, and the troponin I complex of the myosin head [83,84,85]. BNP is an essential marker in heart failure, which is produced by the left ventricle when there is a myocardial stretch. The early detection of BNP is vital to prevent and assess future treatment [86,87,88]. The chronic cardiac toxicity of doxorubicin causes heart failure and particularly reduces fraction ejection. BNP affects increases in vasodilation, diuresis, and natriuresis. As a result, the American Heart Association recommends BNP and NT-proBNP as a biomarkers of heart failure [89,90]. C-reactive proteins (CRP), which are recognized as inflammatory markers, are potentially used to predict the adverse incidence of cardiac injury [91]. In many studies, both in vivo and vitro models of flavonoid-attenuated doxorubicin-cardiotoxicity have used AST, LDH, CK-MB, Troponin, BNP, and CRP as markers to predict the potency of flavonoids (quercetin, rutin luteolin, and apigenin) and emphasize the injury caused by doxorubicin. It has been acknowledged that these biomarkers are continuously used for the analysis of cardiac toxicity, but a more relevant study is needed to conclude on the biomarker that should be used to predict toxicity. Figure 1 shows the mechanism of doxorubicin increasing ROS, mitochondrial dysfunction, ER stress, DNA break, apoptosis, and cardiac markers.

3. Flavonoid

Flavonoids are an essential group of a natural compounds, generally discovered in many plants [92]. Particularly, their pharmacological activities have been tested intensively in animal models, cell models, and human trials [93,94,95]. Some of them found the inhibition of Nf-KB [96], scavenging radicals [97], antiplatelets [98], anti-thrombotic [99], angiotensin-converting enzyme inhibitors [100], anti-carcinogenic [101,102], anti-calcium overload +, lipoxygenase inhibitors [103], etc. Flavonoids are categorized into some subclasses depending on the C ring and B ring, as well as unsaturation and the oxidation of the C ring. These are flavonols (quercetin, rutin, myristine, morin, kaempferol), flavones (apigenin, luteolin), anthocyanin (cyanidin, malvidin), flavanones (hesperetin, naringin, naringenin) and isoflavonoids (genistin, genistein) [104]. The flavonoid structures are shown in Figure 2.

3.1. Luteolin

Luteolin (3,4,5,7-tetrahydroxy flavone) is discovered in many natural resources, such as vegetables and fruits that are used daily in human life [105]. Furthermore, it has been tested in many pharmacological activities, including anticancer, antidiabetic, anti cholesterol, and cardioprotective measures against doxorubicin [106,107]. Moreover, it is known to inhibit the carbonyl reductase 3 for the conversion of doxorubicin into doxorubicinol [108,109]. In Chinese traditional medicine, plants rich in luteolin are used to treat inflammation, hypertension, and to increase the luteolin plays a vital role as an anticancer agent in multiple mechanisms, such as the suppression of kinase, the regulation of cells, and apoptosis. It has shown many positive effects regarding the multiple cardioprotective effects against ischemia/reperfusion, heart failure, and atherosclerosis [110,111,112]. The luteolin experimental study was conducted in both in vitro and in vivo models against doxorubicin-induced cardiotoxicity. Moreover, a study conducted by [113] reported that rats which were administered a cumulative dose of doxorubicin at 16 mg/kg, as well as luteolin of 50 and 100 mg/kg, for the luteolin groups, showed that both doses of 50 and 100 mg/kg attenuated the toxicity of doxorubicin. The cardiac biomarkers such as troponin T, BNP, and LDH in doxorubicin in the co-treatment luteolin group were significantly reduced. Meanwhile, in the other groups, only the rats which were administered doxorubicin significantly increased. Furthermore, luteolin increases the expression of phlpp1 and p-Akt protein. phlpp1 has been known to regulate the AKT protein to increase cell survival, which minimizes the apoptosis caused by doxorubicin. Interestingly, a similar result also reported that in the pretreatment of luteolin 10 and 20 µM on H9c2 induced by DOX, the 10 µM showed a depletion of ROS production. On the other hand, the H9c2 cell that was only given DOX significantly increased the ROS level, while the pretreatment of luteolin increased the expression of PTEN, and decreased ERK, AKT, and mTOR [114]. Recently, many studies have reported that mitochondria are the main target of DOX toxicity and have led to the opening of the mitochondrial permeability transition pore (mPTP) [115]. The PI3K/Akt and ERK play an essential role in cell proliferation, apoptosis, and migration of the cells [116]. Another study implied that DOX activated several downstream pathways, including PI3K/Akt, while the main role of luteolin is to block the phosphorylation of PI3K, which causes the decrease in ERK, AKT, and mTOR [117]. In an in vivo study, CK-MB, LDH, and some specific cardiac biomarkers including BNP, Troponin T, and CRP were used to assess cardiotoxicity rate [118]. An agreement study by Syahputra showed that rats induced by doxorubicin significantly increased their BNP and Troponin T levels [119]. In some studies, the antioxidant was widely determined by the abundance of ROS production. SOD plays the main role in the neutralization of O-, which is radically active in H2O2, and which is less toxic [120]. The previous study stated that doxorubicin reduced the SOD levels while pretreatment of luteolin increased the SOD levels [117]. Luteolin- 7-O-β-D-glucopyranoside isolated from Dracocephalum tanguticum can reduce the production of CK and LDH and inhibit the increase in ROS expression on H9c2-treated Dox [121]. Interestingly, luteolin has anti-calcium overload qualities, whereby Ca2+ plays an essential role in the contraction and relaxation of the cardiac muscle; therefore, it is important to maintain the level of Ca2+ level, since its imbalance could lead to the loss of cardiac function. SERCA2a plays an important role in maintaining the reuptake of this Ca2+. This luteolin significantly increased the SERCA2a expression in rats with an injured myocardium, which prevented contractile impairment [122]. Therefore, this study conducted proper documentation of the contribution of luteolin against doxorubicin-induced cardiotoxicity. Table 1 completely shows the study design and the doses administered for both luteolin and doxorubicin, and the durations and parameters.

3.2. Quercetin

Quercetin is a flavonol group that is generally found in many plants, such as berries, onions, green hot paper, apples, pears, spinach, etc. [123]. Furthermore, its daily intake in humans is estimated to be 20–50 mg [124]. This promising natural compound has been widely tested for numerous pharmacological activities, including anticancer, antidiabetic, anti-analgesic, and anti-inflammatory properties, and as a cardioprotective it encounters multiple causes such as doxorubicin-induced cardiotoxicity, ischemia/reperfusion injury, and diabetic cardiomyopathy [125,126]. Quercetin acts on several upstream and downstream signaling pathways of the cells such as cardiomyocytes, which are beneficial for cell survival. Quercetin, which downregulates the protein of ERK and MAP kinase on cardiac cell injury [127] was tested on H9c2 cells; the results showed that upregulating the expression of the Bmi-1 protein played the main role in ROS generation and mitochondrial function. Bmi-1 modulated the antioxidant defenses by suppressing the p53 pro-oxidant protein [128]. Furthermore, the H9c2 cell treated with quercetin showed a significantly reduced apoptotic effect, while the H9c2 cell that was only administered doxorubicin significantly increased the apoptotic cell. Moreover, in the in vivo models, mice treated with 20 mg/kg of doxorubicin alone significantly reduced in heart weight and heart-to-body weight ratio. The results of this study showed an increase in creatinine kinase (CK) and LDH as cardiac biomarkers in mice treated with only doxorubicin. Meanwhile, the mice treated with 100 mg/kg of quercetin during pretreatment significantly reduced the cardiac biomarkers [128]. The results clearly showed that quercetin counters the production of oxidative stress by doxorubicin. The simultaneous administration of quercetin with resveratrol on H9c2 cells showed a significant reduction in ROS, AST, ALT, and CK [127]. Interestingly, pre-treatment of quercetin with H9c2 elevated the expression of protein 14-3-3γ which is involved in the protection of myocardial injury. Therefore, pretreatment of quercetin suppressed caspase-3 activity, while the H9c2 cell which was administered with doxorubicin alone inclined the caspase-3 activity. The cell pretreatment of quercetin prevented the opening of mPTP, while in the Dox alone, mPTP was high such that it stimulated the swelling of mitochondria and the excessive release of ROS [129]. In agreement with a study reported by [130], the combination of 80 mg/kg quercetin with sitagliptin on a rat induced with a doxorubicin accumulative dose of 18 mg/kg showed a significant reduction of cardiac biomarkers LDH, CK, Troponin, and CRP. Meanwhile, the doxorubicin group showed a significant trend in these parameters. The combination of sitagliptin and quercetin was more potent compared to sitagliptin and quercetin alone against doxorubicin, and multiple mechanisms including antioxidant, anti-inflammatory, and lipid-lowering effects contributed against doxorubicin [130]. In correlation with the Asma study on an in vivo model, the co-administration of a quercetin dose of 10 mg/kg with losartan of 0.7 mg/kg, on rats induced with doxorubicin of 15 mg/kg, inclined with the myocardial antioxidant enzymes such as SOD and CAT, while the marker of oxidative stress MDA declined [131]. Moreover, the TNF alpha, which is already known as a pro-inflammatory cytokine that stimulates ROS, increased in doxorubicin alone. Interestingly, quercetin attenuated the TNF alpha and the Nuclear Factor-Kappa B (NF-κB). This inhibition was mediated by the antioxidant ability of quercetin. The combination of losartan and quercetin showed a better correlation against the toxicity of doxorubicin than losartan and quercetin alone, because of the synergetic effect [132]. An in vivo study stated that quercetin 2 mg/kg/day for 7 days attenuated the cardiac toxicity caused by DOX, which significantly changed the cardiac biomarkers and significantly improved cardiac histology [133].

3.3. Apigenin

Flavonoids are natural compounds in almost all plants tissue. One of them includes apigenin, which belongs to the sub-classes of flavone [134]. Apigenin has several interesting pharmacological activities which include antioxidant, inflammation, autoimmune, neurodegenerative, and antidiabetic effects, etc. [135]. Furthermore, it upregulates the cell signaling pathway PI3K/Akt and downregulates NF-κB, as well as reduces COX-2 expression. Interestingly, apigenin has well documented elevated antioxidant enzymes such as SOD, Catalase, and Glutathione for encountering cellular oxidants (Sahu et al., 2019). Interleukin 6 (IL-6) and the TNF alpha, which are known as pro-inflammatory cytokines, are attenuated by apigenin. In an antidiabetic animal model, apigenin regulated hyperglycemia and neutralized the reactive oxygen species (ROS) [136]. Moreover, it played a vital role in encountering cardiac remodeling, cardiac apoptosis, and toxicity due to doxorubicin [136]. Table 1 below shows a few reports on apigenin against doxorubicin. Apigenin is a common compound discovered in countless plants, and it has strong antioxidant activity. Some studies were conducted on behalf of the cardioprotective activity in both animal and cardiac cell experiments. A study conducted by Zara et al. showed that apigenin increased the body weight and heart body weight relative ratio in rats treated with doxorubicin. Hence, the ejection fraction was significantly improved, and there was no difference with normal rat groups. In addition, the percentage of fibrosis on the cardiac cell was decreased compared to doxorubicin alone [137]. Cardiac biomarker injuries, such as CK-MB, LDH, and troponin T, were decreased in the apigenin + doxorubicin group. The expression of anti-apoptotic protein Bcl was decreased, and the expression of pro-apoptotic caspase 3 and Bax was also decreased [137]. In agreement with the previous study, apigenin significantly reduced myocardial enzymes AST, LDH, and CK [138]. In addition, mitochondrial dysfunction has been recently highlighted as a major incense in rats treated with doxorubicin. Interestingly, the ratio of Bax/bcl2 was increased in the doxorubicin treatment group, although, on the other hand, the apigenin protein pro-apoptotic was reversed. Furthermore, apigenin also activated the PI3K/Akt/mTOR pathway in rats treated with doxorubicin, which increased apoptosis. Apigenin of 75 mg/kg leads to the activation of peroxisome proliferator-activated receptor-γ (PPAR-γ) on the rat-induced myocardial infraction, reducing the CK-MB and LDH, as well as reducing the expression of Bax. The stimulation of PPAR-γ decreased in size and inflammation [136]. A similar study conducted by [139] stated that apigenin attenuated myocardial/reperfusion injury by significantly reducing TNF alpha, phospho-IkB-, NF-κB, and ICAM-1. Interestingly, apigenin depleted the inflammatory biomarker of COX-2 and iNOS expression on rats.

3.4. Rutin

Rutin, or quercetin-3-O-rutinoside, is one of the flavonoid compounds which belongs to the subclasses of flavanol [140]. Furthermore, more than 60 plant species which include Vernonia amygdalina from the family Asteraceae contain rutin [141]. As with many other compounds of flavonoids, a major problem is poor bioavailability due to low solubility, and unstable and poor permeability [142]. Rutin is pharmacologically known to attenuate cardiac remodeling by blocking some cellular signaling pathways and ROS [143]. Doxorubicin is known to cause dilation of the left ventricle, leading to cardiac impartment, specifically reducing the ejection fraction. In addition, it generates ROS, apoptosis, lipid peroxidation, and inflammatory response, while endogenous antioxidant SOD, GSH, Catalase are reduced [144]. Rutin has been tested on several in vivo and in vitro models against doxorubicin, and the data is well documented in Table 1. Rutin has significantly shown a decrease in ROS and apoptosis in H9c2 cells treated with doxorubicin. Meanwhile, groups that were administered with doxorubicin only showed a significant increase in ROS and apoptosis [118]. A previous study stated that the activation of ROS-dependent p38 MAPK and the deactivation of ERK signaling pathway led to myocardial apoptosis by doxorubicin [118]. Interestingly, the existence of rutin in the H9c2 cardiomyoblast cell protects the activation of the p38 MAPK signaling pathway [118]. Moreover, the expression of pro-apoptotic proteins caspase 3, caspase 7, and caspase 9 in H9c2 cells on apigenin treatment show a significant decrease. A similar study conducted by Ma et al. (2016) noted that rutin increased the ejection fraction (%) and fractional shortening (%) in mice induced with doxorubicin. Moreover, the cardiac histopathology showed that the fibrosis area was significantly decreased compared to mice administered with doxorubicin alone [145]. Interestingly, this is in line with the apoptotic cell (%) that significantly decreased in the treatment of apigenin. Furthermore, the expression of Bcl-2 was significantly increased in the H9c2-treated apigenin in the doxorubicin group [146].

3.5. Cyanidin

A study conducted by Sixue et al. showed that purple sweet potato anthocyanin (PSPA) of doses 400, 600, 800 μg/mL significantly increased the cell viability of H9c2 compared to the control group. The NO secretion and TNF-α were significantly decreased in H9c2 treated with DOX 3 μmol/L + PSPA dose 200 and 400 μg/mL. Interestingly, the CK, LDH, and TMAO also significantly decreased in H9c2 which was treated with DOX 3 μmol/L + PSPA dose 200 and 400 μg/mL [147]. Furthermore, the result of the vivo model showed that mice that were treated with PSPA of doses 100 and 200 mg/kg had serum and heart tissue levels of LDH, CK, TNF-α, TMAO that were significantly decreased compared with the doxorubicin group [147]. Additionally, anthocyanin has the ability to reduce the level of pro-inflammatory cytokine, which increases in both H9c2 and mice treated with doxorubicin [147]. This is in line with the result of histological analysis of the heart, which shows that doxorubicin causes myocardial rupture and an excessive amount of inflammatory infiltration. However, mice that were receiving PSPA 200 observed only small inflammatory infiltration. This is in line with a study contributed by [148], which stated that cyanidin-3 glucoside can reduce the toxicity caused by doxorubicin, showing that it increases cell viability and decreases renin-angiotensin-aldosterone (RAA). Anthocyanins also significantly decreases reactive oxygen species such as ROS, O.2, OH., H2O2, ONOO-, NO, and the MDA as a marker of lipid peroxidation on H9c2 cell-induced doxorubicin [149]. Generally, cyanidin inhibits apoptosis, pro-inflammatory cytokines, ROS production, and lipid peroxidation; however, it elevates cell viability and antioxidant effects.

3.6. Hesperidin

Hesperidin is one of the flavonoids subclasses of flavanones, which has been extensively tested for its pharmacological activities both in vitro, in vivo, and in silico. The antioxidant properties of hesperidin are extremely high; this shows that hesperidin has potent scavenging activity with the inhibitory concentration of 10.60 μg/mL [150]. The results of a study conducted by Trivedi et al. show that the hesperetin dose of 100 mg/kg significantly reduced the MDA in rat serum induced with doxorubicin [151]. Additionally, the expression of NF-κB, p38, and caspase on cardiac histopathology by IHC showed a significant decrease in rats receiving hesperidin of 100 mg/kg compared with the group which was only injected with doxorubicin. In addition to hesperidin, a previous study revealed that hesperidin formulated with solid nanoparticles (SLN) decreased the cardiac markers of CK-MB and troponin I on rats treated with doxorubicin [152]. Moreover, the hesperidin-SLN decreased the lipid peroxidation but increased the CAT and SOD levels [152]. A similar study showed that rats induced with doxorubicin and given hesperidin 50 mg/kg (3 times a week) had significantly decreased levels of LDH, CK, NO, MPO, MDA, and Caspase-3, compared with the groups that given only doxorubicin [153].

3.7. Chrysin

Chrysin is a flavonoid discovered in honey, mushrooms, and several plants [154]. Currently, several pharmacological activities of chrysin have been tested on cell and rat models. In cardioprotective activity, chrysin depleted the production of ROS, which caused decreases in protein p38, p53, and Nf-κB in cardiac cells [155]. The activation of the p53 cardiac cell stimulated the protein pro-apoptosis Bax and decreased the protein anti-apoptosis Bcl-2. Doxorubicin plays an important role in increasing PTEN and decreasing VEGF, which downregulates the AKT protein that causes apoptosis [154]. Furthermore, a study conducted by Mantawy showed that chrysin reduced pro-inflammatory markers such as Nf-κB, iNOS, COX-2, and TNF-α on rats intoxicated with doxorubicin. Therefore, Nf-κB plays an essential role in the downstream of the inflammatory response which causes pathological changes in the cardiac cell [156].

3.8. Naringenin and Narigin

Naringenin and naringin are flavonoids that belongs to the isoflavonoid subclasses, widely found in grapes and citrus [157]. Furthermore, they have been tested in some pharmacological activities, including against ischemia/reperfusion and cardioprotective activity against doxorubicin [158]. A conducted study showed that naringenin increased antioxidant endogenous activities such as SOD, GPx, GST, Catalase, and GSH [159]. This antioxidant diminishes superoxide anion, superoxide radical, and hydrogen peroxide that increase while being treated with doxorubicin [160]. Moreover, naringenin is known to increase the expression of Nrf-2 as an antioxidant modulator to produce more antioxidants [161]. Subsequently, the cardiac marker injury was also decreased with the co-treatment of naringenin on rats intoxicated with doxorubicin (CK-MB, LDH, AST, and Troponin T) [162]. Meanwhile, the lipid peroxidation was decreased on a rat receiving naringenin. Moreover, inflammation markers such as TNF-α, IL-6, and IL-10 [162] were decreased compared to the previous study. A summary of the mechanism is presented in Figure 3.
Table 1. The cardioprotective activity of flavonoids against doxorubicin-induced cardiotoxicity.
Table 1. The cardioprotective activity of flavonoids against doxorubicin-induced cardiotoxicity.
CompoundStudy DesignFlavonoid DoseDoxorubicin DoseDurationParametersReferences
LuteolinIn vivo (rat)50 mg/kg
100 mg/kg
(P.O 1 week in advance and gastric administration lasted for 5 weeks)
16 mg/kg
(Intraperitoneal injection once a week)
5 weeks↓BNP, ↓CK-MB, ↓MDA, ↓LDH, ↑SOD, ↑Bcl2, ↓Bax, ↑p-AKT, ↓Caspase-3[48]
Luteolin-7-O-glucoside In vitro (H9c2)10 and 20 µM
(pre-treated for 24 h)
10 µM
(Incubated for 24 h)
48 h↑Cell viability, ↓apoptosis, ↓ROS, ↑P-PTEN, ↓P-Akt, ↓P-ERK, ↓p-mTOR, ↓p-GSK-3bate [114]
LuteolinIn vitro (H9c2)5, 10, 20 µM
(pre-treated for 24 h)
20 µM
(Incubated for 24 h)
48 h↑Cell viability, ↓CK, ↓LDH, ↓ROS, ↓ [Ca2+]i [118]
LuteolinIn vitro (AMCMs)1, 10, 50 µM1 µM24 h↓LDH, ↓CK, ↓Apoptosis, ↓ROS, ↑Bcl-2, ↓Bax, ↓Caspase 9, ↑Bnip3, ↑Parkin, ↑Pink1, ↑LC3BII, ↑P62, ↓mTOR, ↑LAMP1, ↑TFEB, ↑Drp1 [117]
QuercetinIn vivo (rat)10, 25, 50 mg/kg
(P.O for 7 weeks)
2 mg/kg
(Intraperitoneal once a week until 4 weeks)
7 weeks↓Blood pressure, ↓HR, ↓LVEDP, ↑coronary flow, ↑+(dp/dt) max, ↑-(dp/dt) max, ↓CK-MB, ↓LDH, ↓Na+, ↓K+, ↓MDA, ↑GSH, ↑SOD, ↑Catalase, ↑Nrf2 [122]
Quercetin In vivo (rat)2 mg/kg
(P.O for 7 days)
10 mg/kg
(I.V on day 5)
7 days↓AST, ↓LDH, ↑GSH, ↓BUN, ↓Creatinine, ↓TBRAS[133]
QuercetinIn vitro
(H9c2)
100 µM
(pre-treated for 48 h and 96 h)
1 µM48 h and 96 h↑CR inhibition, ↓LDH, ↓iron chealting, ↓LPO IC50 [163]
QuercetinIn vitro
(H9c2)
50 and 100 µM
(Incubated 48 h)
0–16μM
(Incubated 48 h)
48 h↑Cell viability, ↓apoptosis, ↑MMP, ↓ROS, ↑Bmi-1[128]
In vivo
(Mice)
100 mg/kg
(P.O for 10 days)
20 mg/kg
(I.P)
48 h↑LVEF, ↑LVFS, ↓LVEDD, ↓LVESD, ↓LDH, ↓MDA, ↑SOD, ↑Bmi-1
Quercetin polymeric micellesIn vitro
(H9c2)
µM0.01, 0.1, 1 µM48 h↓Caspase 3, ↓caspase 7, ↓ROS, ↓apoptosis[164]
In vivo
(mice)
3.31 mg/kg
(I.V every 3 days for 3 cycle)
6 mg/kg
(I.V every 3 days for 3 cycle)
10 days↓AST, ↓ALT, ↓CK
QuercetinIn vitro
(Neonatal
Rat cardiomyocytes)
10,20,40,80 µM
(pre-treated for 22 h)
1 µM (incubated 24 h)48 h (2 h normal condition)↑Cell viability, ↓LDH, ↓caspase 3, ↓apoptosis, ↑14-3-3γ, ↑MMP, ↑SOD, ↑Catalase, ↑Gpx, ↓MDA, ↑GSH, ↑GSSG[129]
QuercetinIn vivo
(rat)
10 mg/kg
(P.O for 6 weeks)
2.5 mg/kg
(I.P every 2 days for 2 weeks)
6 weeks↓CK-MB, ↓LDH, ↓TNF, ↑SOD, ↑CAT, ↓MDA, ↓NO[131]
Apigenin In vivo (rat)25 mg/kg
(P.O for 12 days)
2 mg/kg
(I.P every 2 days for 12 days)
12 days↑%EF, ↑%FS, ↓LVIDd, ↓LVISd, ↓LDH, ↓CK-MB, ↓cTn-I, ↓ALT, ↓AST, ↓%Fibrosis, ↓MDA, ↑SOD, ↑Catalase, ↑Bcl-2, ↓Bax, ↓Caspase-3[137]
ApigeninIn vitro
(Murine cardiomyocytes)
20 µM
(Incubated for 24 h)
1 µM (incubated for 24 h)24 h↑Cell viability, ↓ROS, TBARS, ↑CAT, ↓Carbonyl protein, ↑SOD, ↑GST, ↑GPx, ↑GSH, ↑GR, ↓DNA fragmentation, ↓8-OHdG, ↓Cyt c, ↑Bcl-2, ↓Bax, ↓caspase 3, ↓caspase 9, ↓caspase 8, Apaf-1, FAS, t-Bid, ↓IκBα, ↓NF-κB, PKC-δ, ↓JNK, ↓p38, ↓p53, ↑PI3K, ↑Akt, mTOR, ↓iNOS, ↑HO-1, and ↑Nrf-2[136]
In vivo (rat)100 mg/kg (P.O 7 days)3 mg/kg (I.P on day 1,3,5)7 days↑Total erythrocytes, ↑Haemoglobin, Total leucocytes, ↓Total cholesterol, HDL, TGD, LDH, ↓CK, ↓AST, ↓Troponin I, ↓Troponin T, ↑SOD, ↓Protein carbonyl, ↓ROS, ↓TBARS, ↑CAT, ↑GPx, ↑GST, ↑GSH, ↓8-OHdG, ↑GR, ↓NADPH oxidase, ↓DNA fragmentation, ↓MMP, ↓Cyt C, ↑Bcl-2, ↓Bax, ↓Caspase 3, ↓caspase 9, ↓caspase 8, ↓FAS, ↓t-Bid, ↓IκBα, ↓NF-κB, ↓PKC-δ, ↓JNK, p38, ↓p53, ↑PI3K, ↑Akt, ↑mTOR, ↓iNOS, ↑HO-1, and ↑Nrf-2
ApigeninIn vivo
(mice)
125 and 250 mg/kg
(Gastric gavage for 17 days)
3 mg/kg
(I.P every 2 days for 16 days)
17 days↓AST, ↓LDH, ↓CK, ↓Apoptosis, ↓Bax, ↑Bcl-2, ↓Beclin1, ↓LC3, ↑p-mTOR, ↑mTOR, ↑p-AKT, ↑AKT1/2/3, ↑PI3K[138]
RutinIn vivo
(H9c2)
10, 30, 50, or 70 μM
(pre-treated for 1 h)
5μM/pirarubicin
(Incubated 24 h)
24 h↑Cell viability, ↓ROS, ↓Apoptosis, ↓caspase 3, ↓caspase 7, ↓caspase 7, TGF-β1, p-p38 MAPK[165]
RutinIn vivo
(mice)
100 mg/kg
(P.O for 11 weeks)
3 mg/kg
(I.P every 2 days for 2 weeks)
11 weeks ↑LVEF, ↑LVFS, ↓%fibrosis, [166]
In vitro
(cardiomyocytes)
10 μM
(pre-treated for 24 h)
1 μM
(incubated for 24 h)
48 h↓Apoptosis, ↑Bcl-2, ↓Caspase 3, ↓P62, ↓LC3BI/II, ↓ATG5
RutinIn vivo
(mice)
100 μmol/kg
(I.P for 5 days)
15 mg/kg
(I.P day 1)
5 days↑GSHpx, ↓MDA, ↓CPK, ↓Total bone marrow, ↓NADPH IC50 [146]
RutinIn vivo (rat)50 mg/kg
(P.O 3 times per week for 3 weeks)
25 mg/kg3 weeks↓Total cholestrol, ↑HDL, ↓LDL, ↓CK, ↓LDH, ↓AST, ↑Glutathione, ↑GPx, ↑Glutathione-s-tranasferase, ↓MDA[167]
HesperidinIn vivo (rat)50 mg/kg
(Gastric administration 3 times per week for 3 weeks)
4 mg/kg
(I.P 3 times per week for 2 weeks)
3 weeks↓CK, ↓LDH, ↓NO, ↓MPO, ↓MDA, ↑GSH, ↑CAT, ↓Caspase 3[168]
AnthocyaninIn vitro (HL-1)0, 5, 25, 125, 250 μM0, 0.125, 0.25, 0.5, 1, 2, 4 μM48 h↑Cell viability, ↓RAS[148]
AnthocyaninIn vitro (H9c2)20 and 40 μg/mL
(post-treated for 24 h)
1 μM
(treated for 6 and 12 h)
36 h↑Cell viability, ↓apoptosis, ↓CHIP, ↑HSF1, ↓IGF-IIR, ↓caspase 3, p-NFκB, ↑p-Akt, ↑ERα, ↑ERβ[149]
ChrysinIn vivo (rat)25 and 50 mg/kg
(P.O for 12 days)
15 mg/kg
(I.P on day 12)
12 days↓CK-MB, ↓LDH, ↓MDA, ↓NF-κB, ↓iNOS, ↓COX-2, ↓Bax, ↑Bcl2, ↓TNF-α, COX-2, ↑SOD, ↑CAT, ↓NO, ↓Apoptosis, ↑GSH, ↑Cyc C[155]
ChrysinIn vivo (rat)50 mg/kg
(P.O 4 times per week for 5 weeks)
5 mg/kg
(I.P once a week for 4 weeks)
4 weeks↓VEGF, ↑AKT, ↑PTEN, ↓NF-κB, ↓Bax, Bcl-2, ↓P53, ↓MAPK, GSH, ↓MDA, ↑CAT, ↑SOD, ↑Gpx, ↑GR[155]
HesperidinIn vivo (rat)25, 50, 100 mg/kg
(P.O 5 times per weeks for 5 weeks)
4 mg/kg
(I.P once a week for 5 weeks)
5 weeks↓MDA, ↑GSH, ↓NF-kB, ↓p38, ↓Caspase-3, ↓apoptosis, ↓% demaged cell[151]
Hesperidin solid nano particle In vivo (rat)20 mg/kg
(P.O for 7 days)
15 mg/kg
(I.P on day 5)
7 days↓CK-MB, ↓Troponin I, ↓MDA, ↑SOD, ↑CAT, ↓Apoptosis, ↓Caspase 3[152]
AnthocyaninIn vitro
(H9c2)
100–800 μg/mL 3 μmol/L for 12 h 12 h↓NO, ↓TNF-α, ↓TMAO, ↓LDH, ↓CK[169]
In vivo
(mice)
100 and 200 mg/kg
(P.O for 25 days)
13 mg/kg injected on day 26, 27, and 1828 days↓NO, ↓LDH, ↓CK, ↓TNF-α ↓TMAO
NaringeninIn vivo (rat)25 mg/kg
(P.O for 7 days)
15 mg/kg
(I.P on day 7)
7 days↓LDH, ↓CPK, ↓MDA, ↑SOD, ↑GSH, ↑CAT, ↑GST[170]
NaringeninIn vivo (rat)100 mg/kg
(P.O for 2 weeks)
15 mg/kg
(I.P on day 14)
2 weeks↓CK-MB, ↓Creatinine, ↓AST, ↓ALT, ↓Urea, ↓LDH, ↓TNF-α, ↓IL-6, ↓IL-1β, ↓TBARS, ↑GSH, ↑CAT, ↑SOD, ↑GST, ↑GPx[160]
naringenin-7-O-glucosideIn vitro (H9c2)5, 10, 20, 40, and 80 μM
(pre-treated for 24 h)
10 μM
(Incubated 24 h)
48 h↓Cell viability, ↓ROS, ↓LDH, ↓CK, ↑GSH, ↑GPx, ↓ [Ca2+]I[171]
NaringeninIn vivo (rat)15 mg/kg
(P.O for 30 days)
15 mg/kg
(I.P on day 30)
30 days↑SOD, ↑CAT, ↑GSH[159]
NaringinIn vivo (rat)50 and 100 mg/kg
(I.P for 14 days)
15 mg/kg
(I.P on day 10)
14 days↑GSH, ↑SOD, ↑CAT, ↓MDA, ↓NADH, ↓Cyt-C, [161]
NaringinIn vivo (rat)50 mg/kg
(P.O for 10 weeks)
3 mg/kg
(I.P on week 1,3,5,7,9)
10 weeks↓LDH, ↓Troponin T, ↓MDA, ↑CAT, ↑SOD, ↑GPx, ↓TGFβ1, ↓TNF-α, ↓IL-6, ↓IL-10 [162]
BNP: brain natriuretic peptide; CK-MB: creatinine kinase-MB; MDA: malondialdehyde; LDH: lactate dehydrogenase; SOD: superoxide dismutase; Bcl2: B-cell lymphome2; TNF-α: tumor necrosis factor; IL: interleukin; ROS: reactive oxygen species; Cty-c: cytochrome c; GPx: glutathione peroxidase; PTEN; phosphatase and tensin homolog; NO: nitrite oxide; MMP: mitochondria membrane potential; GSH: glutathione; CAT: catalase; NADH: nicotinamide adenine dinucleotide; HDL: high density lipoprotein; LDL: low density lipoprotein; RAS: renin angiotensin aldosterone; iNOS: inducible nitrite oxide; LVEF: left ventricular ejection fraction; LVFS: left ventricular ejection shortening; NF-κB: nuclear factor kappa B; HR: heart rate; 8-OHdG: 8-Oxo-2’-deoxyguanosine; CR: carbonyl reductase; HO-1: heme oxygenase-1; nrf-2: Nuclear factor-erythroid factor 2-related factor 2; AST: aspartate transaminase; ALT: alanine transferase; TGFβ1: transforming growth beta 1; MAPK: mitogen activated protein kinase; COX-2: cyclooxygenase; Bnip3: BCL2 interacting protein 3.

4. Conclusions

In conclusion, doxorubicin has multiple mechanisms that cause cardiac toxicity, including decreased antioxidant effects, decreased mitochondrial function, increased lipid peroxidation, and increased inflammatory response. Furthermore, it has been acknowledged that no study elucidates the predominant mechanism. The dietary supplements in flavonoids, such as quercetin, rutin, luteolin, apigenin, hesperidin, anthocyanin, and naringenin, play an essential role in combatting cardiac toxicity by multiple mechanisms which are reducing ROS, lipid peroxidation, mitochondria permeability and the suppress apoptosis (Figure 3). In the future, it is suggested that more mechanism activities of flavonoids against doxorubicin-induced cardiotoxicity are explored.

Author Contributions

Conceptualization, R.A.S. and U.H.; supervision, A.D. and M.P.N.; funding acquisition, D.S. All authors have read and agreed to the published version of the manuscript.

Funding

This study supported by Sandwich/PKPI Program 2021 and Hibah PMSU Tahun III (12/E1/KP.PTNBH/2021) from Kementerian Pendidikan dan kebudayaan, Indonesia.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors thankful to Simona Saponara from University of Siena, Italy for the feedback for this manuscript.

Conflicts of Interest

The authors declare there are no conflict interest.

References

  1. Van der Zanden, S.Y.; Qiao, X.; Neefjes, J. New insights into the activities and toxicities of the old anticancer drug doxorubicin. FEBS J. 2021, 288, 6095–6111. [Google Scholar] [CrossRef] [PubMed]
  2. Li, D.; Yang, Y.; Wang, S.; He, X.; Liu, M.; Bai, B.; Tian, C.; Sun, R.; Yu, T.; Chu, X. Role of acetylation in doxorubicin-induced cardiotoxicity. Redox Biol. 2021, 46, 102089. [Google Scholar] [CrossRef]
  3. De Oliveira, B.L.; Niederer, S. A biophysical systems approach to identifying the pathways of acute and chronic doxorubicin mitochondrial cardiotoxicity. PLoS Comput. Biol. 2016, 12, e1005214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Cardinale, D.; Colombo, A.; Bacchiani, G.; Tedeschi, I.; Meroni, C.A.; Veglia, F.; Civelli, M.; Lamantia, G.; Colombo, N.; Curigliano, G.; et al. Early detection of anthracycline cardiotoxicity and improvement with heart failure therapy. Circulation 2015, 131, 1981–1988. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Bernstein, D. Anthracycline Cardiotoxicity: Worrisome Enough to Have You Quaking? Circ. Res. 2018, 122, 188–190. [Google Scholar] [CrossRef] [PubMed]
  6. Lefrak, E.A.; Piťha, J.; Rosenheim, S.; Gottlieb, J.A. A clinicopathologic analysis of adriamycin cardiotoxicity. Cancer 1973, 32, 302–314. [Google Scholar] [CrossRef]
  7. Reinbolt, R.E.; Patel, R.; Pan, X.; Timmers, C.D.; Pilarski, R.; Shapiro, C.L.; Lustberg, M.B. Risk factors for anthracycline-associated cardiotoxicity. Support. Care Cancer 2016, 24, 2173–2180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Armenian, S.H.; Lacchetti, C.; Barac, A.; Carver, J.; Constine, L.S.; Denduluri, N.; Dent, S.; Douglas, P.S.; Durand, J.B.; Ewer, M.; et al. Prevention and monitoring of cardiac dysfunction in survivors of adult cancers: American Society of Clinical Oncology Clinical Practice Guideline. J. Clin. Oncol. 2017, 35, 893–911. [Google Scholar] [CrossRef] [PubMed]
  9. Johnson, C.B.; Davis, M.K.; Law, A.; Sulpher, J. Shared risk factors for cardiovascular disease and cancer: Implications for preventive health and clinical care in oncology patients. Can. J. Cardiol. 2016, 32, 900–907. [Google Scholar] [CrossRef]
  10. Songbo, M.; Lang, H.; Xinyong, C.; Bin, X.; Ping, Z.; Liang, S. Oxidative stress injury in doxorubicin-induced cardiotoxicity. Toxicol. Lett. 2019, 307, 41–48. [Google Scholar] [CrossRef]
  11. Hasinoff, B.B.; Patel, D.; Wu, X. The role of topoisomerase IIβ in the mechanisms of action of the doxorubicin cardioprotective agent dexrazoxane. Cardiovasc. Toxicol. 2020, 20, 312–320. [Google Scholar] [CrossRef] [PubMed]
  12. He, H.; Wang, L.; Qiao, Y.; Zhou, Q.; Li, H.; Chen, S.; Yin, D.; Huang, Q.; He, M. Doxorubicin induces endotheliotoxicity and mitochondrial dysfunction via ROS/eNOS/NO pathway. Front. Pharmacol. 2020, 10, 1531. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Agustini, F.D.; Arozal, W.; Louisa, M.; Siswanto, S.; Soetikno, V.; Nafrialdi, N.; Suyatna, F. Cardioprotection mechanism of mangiferin on doxorubicin-induced rats: Focus on intracellular calcium regulation. Pharm. Biol. 2016, 54, 1289–1297. [Google Scholar] [CrossRef]
  14. Qin, Y.; Guo, T.; Wang, Z.; Zhao, Y. The role of iron in doxorubicin-induced cardiotoxicity: Recent advances and implication for drug delivery. J. Mater. Chem. B 2021, 9, 4793–4803. [Google Scholar] [CrossRef] [PubMed]
  15. Zhao, L.; Zhang, B. Doxorubicin induces cardiotoxicity through upregulation of death receptors mediated apoptosis in cardiomyocytes. Sci. Rep. 2017, 7, 44735. [Google Scholar] [CrossRef] [Green Version]
  16. Shabalala, S.; Muller, C.J.; Louw, J.; Johnson, R. Polyphenols, autophagy and doxorubicin-induced cardiotoxicity. Life Sci. 2017, 180, 160–170. [Google Scholar] [CrossRef] [PubMed]
  17. Chen, B.; Zhong, L.; Roush, S.F.; Pentassuglia, L.; Peng, X.; Samaras, S.; Davidson, J.M.; Sawyer, D.B.; Lim, C.C. Disruption of a GATA4/Ankrd1 signaling axis in cardiomyocytes leads to sarcomere disarray: Implications for anthracycline cardiomyopathy. PLoS ONE 2012, 7, e35743. [Google Scholar] [CrossRef] [Green Version]
  18. Vejpongsa, P.; Yeh, E.T. Prevention of anthracycline-induced cardiotoxicity: Challenges and opportunities. J. Am. Coll. Cardiol. 2014, 64, 938–945. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Gyöngyösi, M.; Lukovic, D.; Zlabinger, K.; Spannbauer, A.; Gugerell, A.; Pavo, N.; Traxler, D.; Pils, D.; Maurer, G.; Jakab, A.; et al. Liposomal doxorubicin attenuates cardiotoxicity via induction of interferon-related DNA damage resistance. Cardiovasc. Res. 2020, 116, 970–982. [Google Scholar] [CrossRef]
  20. Reichardt, P.; Tabone, M.D.; Mora, J.; Morland, B.; Jones, R.L. Risk–benefit of dexrazoxane for preventing anthracycline-related cardiotoxicity: Re-evaluating the European labeling. Future Oncol. 2018, 14, 2663–2676. [Google Scholar] [CrossRef] [PubMed]
  21. Sawaya, H.; Sebag, I.A.; Plana, J.C.; Januzzi, J.L.; Ky, B.; Cohen, V.; Gosavi, S.; Carver, J.R.; Wiegers, S.E.; Martin, R.P.; et al. Early detection and prediction of cardiotoxicity in chemotherapy-treated patients. Am. J. Cardiol. 2011, 107, 1375–1380. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Dillard, C.J.; German, J.B. Phytochemicals: Nutraceuticals and human health. J. Sci. Food Agric. 2000, 80, 1744–1756. [Google Scholar] [CrossRef]
  23. Sadzuka, Y.; Sugiyama, T.; Shimoi, K.; Kinae, N.; Hirota, S. Protective effect of flavonoids on doxorubicin-induced cardiotoxicity. Toxicol. Lett. 1997, 92, 1–7. [Google Scholar] [CrossRef]
  24. Bast, A.; Kaiserová, H.; Den Hartog, G.J.; Haenen, G.R.; Van Der Vijgh, W.J. Protectors against doxorubicin-induced cardiotoxicity: Flavonoids. Cell Biol. Toxicol. 2007, 23, 39–47. [Google Scholar] [CrossRef]
  25. Feliciano, R.P.; Pritzel, S.; Heiss, C.; Rodriguez-Mateos, A. Flavonoid intake and cardiovascular disease risk. Curr. Opin. Food Sci. 2015, 2, 92–99. [Google Scholar] [CrossRef]
  26. Bartlett, J.J.; Trivedi, P.C.; Pulinilkunnil, T. Autophagic dysregulation in doxorubicin cardiomyopathy. J. Mol. Cell. Cardiol. 2017, 104, 1–8. [Google Scholar] [CrossRef]
  27. Alkuraishy, H.M.; Al-Gareeb, A.I.; Al-hussaniy, H.A. Doxorubicin-induced cardiotoxicity: Molecular mechanism and protection by conventional drugs and natural products. Int. J. Clin. Oncol. Cancer Res. 2017, 2, 31–44. [Google Scholar]
  28. Pilco-Ferreto, N. and Calaf, G.M. Influence of doxorubicin on apoptosis and oxidative stress in breast cancer cell lines. Int. J. Oncol. 2016, 49, 753–762. [Google Scholar] [CrossRef] [Green Version]
  29. Carvalho, F.S.; Burgeiro, A.; Garcia, R.; Moreno, A.J.; Carvalho, R.A.; Oliveira, P.J. Doxorubicin-induced cardiotoxicity: From bioenergetic failure and cell death to cardiomyopathy. Med. Res. Rev. 2014, 34, 106–135. [Google Scholar] [CrossRef]
  30. Yan, Y.; Finkel, T. Autophagy as a regulator of cardiovascular redox homeostasis. Free Radic. Biol. Med. 2017, 109, 108–113. [Google Scholar] [CrossRef] [PubMed]
  31. Ma, J.; Wang, Y.; Zheng, D.; Wei, M.; Xu, H.; Peng, T. Rac1 signalling mediates doxorubicin-induced cardiotoxicity through both reactive oxygen species-dependent and -independent pathways. Cardiovasc. Res. 2013, 97, 77–87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Kalivendi, S.V.; Kotamraju, S.; Zhao, H.; Joseph, J.; Kalyanaraman, B. Doxorubicin-induced apoptosis is associated with increased transcription of endothelial nitric-oxide synthase: Effect of antiapoptotic antioxidants and calcium. J. Biol. Chem. 2001, 276, 47266–47276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Beinert, H.; Kennedy, M.C.; Stout, C.D. Aconitase as iron-sulfur protein, enzyme, and iron-regulatory protein. Chem. Rev. 1996, 96, 2335–2374. [Google Scholar] [CrossRef] [PubMed]
  34. Panjrath, G.S.; Patel, V.; Valdiviezo, C.I.; Narula, N.; Narula, J.; Jain, D. Potentiation of doxorubicin cardiotoxicity by iron loading in a rodent model. J. Am. Coll. Cardiol. 2007, 49, 2457–2464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Mitry, M.A.; Edwards, J.G. Doxorubicin induced heart failure: Phenotype and molecular mechanisms. IJC Heart Vasc. 2016, 10, 17–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Octavia, Y.; Tocchetti, C.G.; Gabrielson, K.L.; Janssens, S.; Crijns, H.J.; Moens, A.L. Doxorubicin-induced cardiomyopathy: From molecular mechanisms to therapeutic strategies. J. Mol. Cell. Cardiol. 2012, 52, 1213–1225. [Google Scholar] [CrossRef] [Green Version]
  37. Gilleron, M.; Marechal, X.; Montaigne, D.; Franczak, J.; Neviere, R.; Lancel, S. NADPH oxidases participate to doxorubicin-induced cardiac myocyte apoptosis. Biochem. Biophys. Res. Commun. 2009, 388, 727–731. [Google Scholar] [CrossRef]
  38. Šimůnek, T.; Štěrba, M.; Popelová, O.; Adamcová, M.; Hrdina, R.; Geršl, V. Anthracycline-induced cardiotoxicity: Overview of studies examining the roles of oxidative stress and free cellular iron. Pharmacol. Rep. 2009, 61, 154–171. [Google Scholar] [CrossRef]
  39. Spallarossa, P.; Garibaldi, S.; Altieri, P.; Fabbi, P.; Manca, V.; Nasti, S.; Rossettin, P.; Ghigliotti, G.; Ballestrero, A.; Patrone, F.; et al. Carvedilol prevents doxorubicin-induced free radical release and apoptosis in cardiomyocytes in vitro. J. Mol. Cell. Cardiol. 2004, 37, 837–846. [Google Scholar] [CrossRef]
  40. Dorn, G.W., II. Mitochondrial dynamics in heart disease. Biochim. Biophys. Acta—Mol. Cell Res. 2013, 1833, 233–241. [Google Scholar] [CrossRef] [Green Version]
  41. Dos Santos, D.S.; dos Santos Goldenberg, R.C. Doxorubicin-induced cardiotoxicity: From mechanisms to development of efficient therapy. In Cardiotoxicity; Tan, W., Ed.; IntechOpen: London, UK, 2018; pp. 3–24. [Google Scholar]
  42. Heller, B.I.; Jacobson, W.E. Renal hemodynamics in heart disease. Am. Heart J. 1950, 39, 188–204. [Google Scholar] [CrossRef]
  43. Guven, C.; Sevgiler, Y.; Taskin, E. Mitochondrial dysfunction associated with doxorubicin. In Mitochondrial Diseases; Taskin, E., Guven, C., Sevgiler, Y., Eds.; IntechOpen: London, UK, 2018; p. 323. [Google Scholar]
  44. Halliwell, B.; Gutteridge, J.M. The definition and measurement of antioxidants in biological systems. Free Radic. Biol. Med. 1995, 18, 125–126. [Google Scholar] [CrossRef]
  45. Dudek, J.; Hartmann, M.; Rehling, P. The role of mitochondrial cardiolipin in heart function and its implication in cardiac disease. Biochim. Biophys. Acta—Mol. Basis Dis. 2019, 1865, 810–821. [Google Scholar] [CrossRef]
  46. Gorini, S.; De Angelis, A.; Berrino, L.; Malara, N.; Rosano, G.; Ferraro, E. Corrigendum to “Chemotherapeutic Drugs and Mitochondrial Dysfunction: Focus on Doxorubicin, Trastuzumab, and Sunitinib”. Oxid. Med. Cell. Longev. 2019, 2019, 9601435. [Google Scholar] [CrossRef] [PubMed]
  47. Ichikawa, Y.; Ghanefar, M.; Bayeva, M.; Wu, R.; Khechaduri, A.; Prasad, S.V.; Mutharasan, R.K.; Naik, T.J.; Ardehali, H. Cardiotoxicity of doxorubicin is mediated through mitochondrial iron accumulation. J. Clin. Investig. 2014, 124, 617–630. [Google Scholar] [CrossRef] [Green Version]
  48. Feijen, E.A.; Leisenring, W.M.; Stratton, K.L.; Ness, K.K.; Van Der Pal, H.J.; Van Dalen, E.C.; Armstrong, G.T.; Aune, G.J.; Green, D.M.; Hudson, M.M.; et al. Derivation of anthracycline and anthraquinone equivalence ratios to doxorubicin for late-onset cardiotoxicity. JAMA Oncol. 2019, 5, 864–871. [Google Scholar] [CrossRef]
  49. Zhang, Q.L.; Yang, J.J.; Zhang, H.S. Carvedilol (CAR) combined with carnosic acid (CAA) attenuates doxorubicin-induced cardiotoxicity by suppressing excessive oxidative stress, inflammation, apoptosis and autophagy. Biomed. Pharmacother. 2019, 109, 71–83. [Google Scholar] [CrossRef] [PubMed]
  50. Birari, L.; Wagh, S.; Patil, K.R.; Mahajan, U.B.; Unger, B.; Belemkar, S.; Goyal, S.N.; Ojha, S.; Patil, C.R. Aloin alleviates doxorubicin-induced cardiotoxicity in rats by abrogating oxidative stress and pro-inflammatory cytokines. Cancer Chemother. Pharmacol. 2020, 86, 419–426. [Google Scholar] [CrossRef] [PubMed]
  51. Yarmohammadi, F.; Rezaee, R.; Karimi, G. Natural compounds against doxorubicin-induced cardiotoxicity: A review on the involvement of Nrf2/ARE signaling pathway. Phytother. Res. 2021, 35, 1163–1175. [Google Scholar] [CrossRef] [PubMed]
  52. Abo-Salem, O.M. The protective effect of aminoguanidine on doxorubicin-induced nephropathy in rats. J. Biochem. Mol. Toxicol. 2012, 26, 1–9. [Google Scholar] [CrossRef] [PubMed]
  53. Li, B.; Kim, D.S.; Yadav, R.K.; Kim, H.R.; Chae, H.J. Sulforaphane prevents doxorubicin-induced oxidative stress and cell death in rat H9c2 cells. Int. J. Mol. Med. 2015, 36, 53–64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Zhao, Y.Q.; Zhang, L.; Zhao, G.X.; Chen, Y.; Sun, K.L.; Wang, B. Fucoxanthin attenuates doxorubicin-induced cardiotoxicity via anti-oxidant and anti-apoptotic mechanisms associated with p38, JNK and p53 pathways. J. Funct. Foods 2019, 62, 103542. [Google Scholar] [CrossRef]
  55. Zhang, Y.; Ahmad, K.A.; Khan, F.U.; Yan, S.; Ihsan, A.U.; Ding, Q. Chitosan oligosaccharides prevent doxorubicin-induced oxidative stress and cardiac apoptosis through activating p38 and JNK MAPK mediated Nrf2/ARE pathway. Chem.-Biol. Interact. 2019, 305, 54–65. [Google Scholar] [CrossRef] [PubMed]
  56. El Btaouri, H.; Morjani, H.; Greffe, Y.; Charpentier, E.; Martiny, L. Role of JNK/ATF-2 pathway in inhibition of thrombospondin-1 (TSP-1) expression and apoptosis mediated by doxorubicin and camptothecin in FTC-133 cells. Biochim. Biophys. Acta—Mol. Cell Res. 2011, 1813, 695–703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Tocchetti, C.; Molinaro, M.; Angelone, T.; Lionetti, V.; Madonna, R.; Mangiacapra, F.; Moccia, F.; Penna, C.; Sartiani, L.; Quaini, F.; et al. Nitroso-redox balance and modulation of basal myocardial function: An update from the Italian Society of Cardiovascular Research (SIRC). Curr. Drug Targets 2015, 16, 895–903. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Valko, M.; Rhodes, C.J.; Moncol, J.; Izakovic, M.M.; Mazur, M. Free radicals, metals and antioxidants in oxidative stress-induced cancer. Chem.-Biol. Interact. 2006, 160, 1–40. [Google Scholar] [CrossRef] [PubMed]
  59. Revis, N.W.; Marušić, N. Effects of doxorubicin and its aglycone metabolite on calcium sequestration by rabbit heart, liver, and kidney mitochondria. Life Sci. 1979, 25, 1055–1063. [Google Scholar] [CrossRef]
  60. Sag, C.M.; Köhler, A.C.; Anderson, M.E.; Backs, J.; Maier, L.S. CaMKII-dependent SR Ca leak contributes to doxorubicin-induced impaired Ca handling in isolated cardiac myocytes. J. Mol. Cell. Cardiol. 2011, 51, 749–759. [Google Scholar] [CrossRef] [Green Version]
  61. Jean, S.R.; Ahmed, M.; Lei, E.K.; Wisnovsky, S.P.; Kelley, S.O. Peptide-Mediated Delivery of Chemical Probes and Therapeutics to Mitochondria. Acc Chem Res. 2016, 49, 1893–1902. [Google Scholar] [CrossRef]
  62. Cui, N.; Wu, F.; Lu, W.J.; Bai, R.; Ke, B.; Liu, T.; Li, L.; Lan, F.; Cui, M. Doxorubicin-induced cardiotoxicity is maturation dependent due to the shift from topoisomerase IIα to IIβ in human stem cell-derived cardiomyocytes. J. Cell. Mol. Med. 2019, 23, 4627–4639. [Google Scholar] [CrossRef] [Green Version]
  63. Rodrigo, R.S.; Nathalie, A.; Elodie, T.; Gonzalo, G.A.; Philippe, T.; Françoise, D.; Julien, D.; Angela, C.; Bérénice, B.; Jean-Yves, B.; et al. Topoisomerase II-alpha protein expression and histological response following doxorubicin-based induction chemotherapy predict survival of locally advanced soft tissues sarcomas. Eur. J. Cancer 2011, 47, 1319–1327. [Google Scholar] [CrossRef] [PubMed]
  64. Marinello, J.; Delcuratolo, M.; Capranico, G. Anthracyclines as topoisomerase II poisons: From early studies to new perspectives. Int. J. Mol. Sci. 2018, 19, 3480. [Google Scholar] [CrossRef] [Green Version]
  65. Yang, F.; Teves, S.S.; Kemp, C.J.; Henikoff, S. Doxorubicin, DNA torsion, and chromatin dynamics. Biochim. Biophys. Acta—Rev. Cancer 2014, 1845, 84–89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Chamberlain, G.R.; Tulumello, D.V.; Kelley, S.O. Targeted delivery of doxorubicin to mitochondria. ACS Chem. Biol. 2013, 8, 1389–1395. [Google Scholar] [CrossRef]
  67. Timm, K.N.; Tyler, D.J. The role of AMPK activation for cardio protection in doxorubicin-induced cardiotoxicity. Cardiovasc. Drugs Ther. 2020, 34, 255–269. [Google Scholar]
  68. Fouad, A.A.; Albuali, W.H.; Al-Mulhim, A.S.; Jresat, I. Cardioprotective effect of cannabidiol in rats exposed to doxorubicin toxicity. Environ. Toxicol. Pharmacol. 2013, 36, 347–357. [Google Scholar] [CrossRef] [PubMed]
  69. Hanna, A.D.; Lam, A.; Tham, S.; Dulhunty, A.F.; Beard, N.A. Adverse effects of doxorubicin and its metabolic product on cardiac RyR2 and SERCA2A. Mol. Pharmacol. 2014, 86, 438–449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Mattila, M.; Koskenvuo, J.; Söderström, M.; Eerola, K.; Savontaus, M. Intramyocardial injection of SERCA2a-expressing lentivirus improves myocardial function in doxorubicin-induced heart failure. J. Gene Med. 2016, 18, 124–133. [Google Scholar] [CrossRef] [PubMed]
  71. Shati, A.A.; Dallak, M. Acylated ghrelin protects the hearts of rats from doxorubicin-induced Fas/FasL apoptosis by stimulating SERCA2a mediated by activation of PKA and Akt. Cardiovasc. Toxicol. 2019, 19, 529–547. [Google Scholar] [CrossRef] [PubMed]
  72. Montaigne, D.; Marechal, X.; Preau, S.; Baccouch, R.; Modine, T.; Fayad, G.; Lancel, S.; Neviere, R. Doxorubicin induces mitochondrial permeability transition and contractile dysfunction in the human myocardium. Mitochondrion 2011, 11, 22–26. [Google Scholar] [CrossRef]
  73. Tocchetti, C.G.; Carpi, A.; Coppola, C.; Quintavalle, C.; Rea, D.; Campesan, M.; Arcari, A.; Piscopo, G.; Cipresso, C.; Monti, M.G.; et al. Ranolazine protects from doxorubicin-induced oxidative stress and cardiac dysfunction. Eur. J. Heart Fail. 2014, 16, 358–366. [Google Scholar] [CrossRef]
  74. Asensio-López, M.C.; Soler, F.; Sánchez-Más, J.; Pascual-Figal, D.; Fernández-Belda, F.; Lax, A. Early oxidative damage induced by doxorubicin: Source of production, protection by GKT137831 and effect on Ca2+ transporters in HL-1 cardiomyocytes. Arch. Biochem. Biophys. 2016, 594, 26–36. [Google Scholar] [CrossRef]
  75. Upadhyay, S.; Gupta, K.B.; Mantha, A.K.; Dhiman, M. A short review: Doxorubicin and its effect on cardiac proteins. J. Cell. Biochem. 2021, 122, 153–165. [Google Scholar] [CrossRef] [PubMed]
  76. Garg, P.; Morris, P.; Fazlanie, A.L.; Vijayan, S.; Dancso, B.; Dastidar, A.G.; Plein, S.; Mueller, C.; Haaf, P. Cardiac biomarkers of the acute coronary syndrome: From history to high-sensitivity cardiac troponin. Intern. Emerg. Med. 2017, 12, 147–155. [Google Scholar] [CrossRef] [Green Version]
  77. Ladenson, J.H. Reflections on the evolution of cardiac biomarkers. Clin. Chem. 2012, 58, 21–24. [Google Scholar] [CrossRef] [Green Version]
  78. Allahham, M.; Singh, M.; Jneid, H. Cardiac Biomarkers in Acute Myocardial Infarction. In Biomarkers in Cardiovascular Disease; Elsevier: Amsterdam, The Netherlands, 2019; pp. 109–114. [Google Scholar]
  79. Ahmad, M.I.; Sharma, N. Biomarkers in acute myocardial infarction. J. Clin. Exp. Cardiol. 2012, 3, 222. [Google Scholar] [CrossRef] [Green Version]
  80. Aldous, S.J. Cardiac biomarkers in acute myocardial infarction. Int. J. Cardiol. 2013, 164, 282–294. [Google Scholar] [CrossRef]
  81. Zhang, G.J.; Luo, Z.H.; Huang, M.J.; Ang, J.J.; Kang, T.G.; Ji, H. An integrated chip for rapid, sensitive, and multiplexed detection of cardiac biomarkers from fingerprick blood. Biosens. Bioelectron. 2011, 28, 459–463. [Google Scholar] [CrossRef]
  82. Fathil, M.F.; Arshad, M.M.; Gopinath, S.C.; Hashim, U.; Adzhri, R.; Ayub, R.M.; Ruslinda, A.R.; Nuzaihan, M.; Azman, A.H.; Zaki, M.; et al. Diagnostics on acute myocardial infarction: Cardiac troponin biomarkers. Biosens. Bioelectron. 2015, 70, 209–220. [Google Scholar] [CrossRef]
  83. Bjurman, C.; Petzold, M.; Venge, P.; Farbemo, J.; Fu, M.L.; Hammarsten, O. High-sensitive cardiac troponin, NT-proBNP, hFABP and copeptin levels in relation to glomerular filtration rates and a medical record of cardiovascular disease. Clin. Biochem. 2015, 48, 302–307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Atas, E.; Kismet, E.; Kesik, V.; Karaoglu, B.; Aydemir, G.; Korkmazer, N.; Demirkaya, E.; Karslioglu, Y.; Yurttutan, N.; Unay, B.; et al. Cardiac troponin-I, brain natriuretic peptide and endothelin-1 levels in a rat model of doxorubicin-induced cardiac injury. J. Cancer Res. Ther. 2015, 11, 882. [Google Scholar] [CrossRef]
  85. Reagan, W.J.; York, M.; Berridge, B.; Schultze, E.; Walker, D.; Pettit, S. Comparison of cardiac troponin I and T, including the evaluation of an ultrasensitive assay, as indicators of doxorubicin-induced cardiotoxicity. Toxicol. Pathol. 2013, 41, 1146–1158. [Google Scholar] [CrossRef] [PubMed]
  86. Teixeira, R.P.; Neves, A.L.; Guimarães, H. Cardiac Biomarkers in Neonatology: BNP/NT pro-BNP, Troponin I/T, CKMB and Myoglobin, a systematic review. J. Pediatr. Neonatal Individ. Med. 2017, 6, e060219. [Google Scholar]
  87. Khiati, S.; Dalla Rosa, I.; Sourbier, C.; Ma, X.; Rao, V.A.; Neckers, L.M.; Zhang, H.; Pommier, Y. Mitochondrial topoisomerase I (top1mt) is a novel limiting factor of doxorubicin cardiotoxicity. Clin. Cancer Res. 2014, 20, 4873–4881. [Google Scholar] [CrossRef] [Green Version]
  88. Johnson, N.A.; Slack, G.W.; Savage, K.J.; Connors, J.M.; Ben-Neriah, S.; Rogic, S.; Scott, D.W.; Tan, K.L.; Steidl, C.; Sehn, L.H.; et al. Concurrent expression of MYC and BCL2 in diffuse large B-cell lymphoma treated with rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone. J. Clin. Oncol. 2012, 30, 3452–3459. [Google Scholar] [CrossRef] [PubMed]
  89. Santaguida, P.L.; Don-Wauchope, A.C.; Oremus, M.; McKelvie, R.; Ali, U.; Hill, S.A.; Balion, C.; Booth, R.A.; Brown, J.A.; Bustamam, A.; et al. BNP, and NT-proBNP as prognostic markers in persons with acute decompensated heart failure: A systematic review. Heart Fail. Rev. 2014, 19, 453–470. [Google Scholar] [CrossRef]
  90. Farnsworth, C.W.; Bailey, A.L.; Jaffe, A.S.; Scott, M.G. Diagnostic concordance between NT-proBNP and BNP for suspected heart failure. Clin. Biochem. 2018, 59, 50–55. [Google Scholar] [CrossRef]
  91. Fertin, M.; Hennache, B.; Hamon, M.; Ennezat, P.V.; Biausque, F.; Elkohen, M.; Nugue, O.; Tricot, O.; Lamblin, N.; Pinet, F.; et al. Usefulness of serial assessment of B-type natriuretic peptide, troponin I, and C-reactive protein to predict left ventricular remodeling after acute myocardial infarction (from the REVE-2 study). Am. J. Cardiol. 2010, 106, 1410–1416. [Google Scholar] [CrossRef]
  92. Terahara, N. Flavonoids in foods: A review. Nat. Prod. Commun. 2015, 10, 521–528. [Google Scholar] [CrossRef] [Green Version]
  93. Agrawal, A.D. Pharmacological activities of flavonoids: A review. Int. J. Pharm. Sci. Nanotechnol. 2011, 4, 1394–1398. [Google Scholar] [CrossRef]
  94. Mulvihill, E.E.; Huff, M.W. Antiatherogenic properties of flavonoids: Implications for cardiovascular health. Can. J. Cardiol. 2010, 26, 17A–21A. [Google Scholar] [CrossRef]
  95. Chiva-Blanch, G.; Badimon, L. Effects of polyphenol intake on the metabolic syndrome: Current evidence from human trials. Oxid. Med. Cell. Longev. 2017, 2017, 5812401. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Serafini, M.; Peluso, I.; Raguzzini, A. Flavonoids as anti-inflammatory agents. Proc. Nutr. Soc. 2010, 69, 273–278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Treml, J.; Šmejkal, K. Flavonoids as potent scavengers of hydroxyl radicals. Compr. Rev. Food Sci. Food Saf. 2016, 15, 720–738. [Google Scholar] [CrossRef]
  98. Zaragozá, C.; Monserrat, J.; Mantecón, C.; Villaescusa, L.; Zaragozá, F.; Álvarez-Mon, M. Antiplatelet activity of flavonoid and coumarin drugs. Vasc. Pharmacol. 2016, 87, 139–149. [Google Scholar] [CrossRef]
  99. Vazhappilly, C.G.; Ansari, S.A.; Al-Jaleeli, R.; Al-Azawi, A.M.; Ramadan, W.S.; Menon, V.; Hodeify, R.; Siddiqui, S.S.; Merheb, M.; Matar, R.; et al. Role of flavonoids in thrombotic, cardiovascular, and inflammatory diseases. Inflammopharmacology 2019, 27, 863–869. [Google Scholar] [CrossRef]
  100. Ojeda, D.; Jiménez-Ferrer, E.; Zamilpa, A.; Herrera-Arellano, A.; Tortoriello, J.; Alvarez, L. Inhibition of angiotensin-converting enzyme (ACE) activity by the anthocyanins delphinidin-and cyanidin-3-O-sambubiosides from Hibiscus sabdariffa. J. Ethnopharmacol. 2010, 127, 7–10. [Google Scholar] [CrossRef]
  101. Mishra, P.K.; Raghuram, G.V.; Bhargava, A.; Ahirwar, A.; Samarth, R.; Upadhyaya, R.; Jain, S.K.; Pathak, N. In vitro and in vivo evaluation of the anticarcinogenic and cancer chemopreventive potential of a flavonoid-rich fraction from a traditional Indian herb Selaginella bryopteris. Br. J. Nutr. 2011, 106, 1154–1168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Luo, Y.; Shang, P.; Li, D. Luteolin: A flavonoid that has multiple cardioprotective effects and its molecular mechanisms. Front. Pharmacol. 2017, 8, 692. [Google Scholar] [CrossRef] [Green Version]
  103. Putri, N.L.; Elya, B.; Puspitasari, N. Antioxidant activity and lipoxygenase inhibition test with total flavonoid content from Garcinia kydia Roxburgh leaves extract. Pharmacogn. J. 2017, 9, 280–284. [Google Scholar] [CrossRef] [Green Version]
  104. Brodowska, K.M. Natural flavonoids: Classification, potential role, and application of flavonoid analogues. Eur. J. Biol. Res. 2017, 7, 108–123. [Google Scholar]
  105. Manzoor, M.F.; Ahmad, N.; Ahmed, Z.; Siddique, R.; Zeng, X.A.; Rahaman, A.; Muhammad Aadil, R.; Wahab, A. Novel extraction techniques and pharmaceutical activities of luteolin and its derivatives. J. Food Biochem. 2019, 43, e12974. [Google Scholar] [CrossRef]
  106. Aziz, N.; Kim, M.Y.; Cho, J.Y. Anti-inflammatory effects of luteolin: A review of in vitro, in vivo, and in silico studies. J. Ethnopharmacol. 2018, 225, 342–358. [Google Scholar] [CrossRef] [PubMed]
  107. Xu, H.; Linn, B.S.; Zhang, Y.; Ren, J. A review on the antioxidative and prooxidative properties of luteolin. React. Oxyg. Species 2019, 7, 136–147. [Google Scholar] [CrossRef] [Green Version]
  108. Arai, Y.; Endo, S.; Miyagi, N.; Abe, N.; Miura, T.; Nishinaka, T.; Terada, T.; Oyama, M.; Goda, H.; El-Kabbani, O.; et al. Structure-activity relationship of flavonoids as potent inhibitors of carbonyl reductase 1 (CBR1). Fitoterapia 2015, 101, 51–56. [Google Scholar] [CrossRef] [PubMed]
  109. Schaupp, C.M.; White, C.C.; Merrill, G.F.; Kavanagh, T.J. Metabolism of doxorubicin to the cardiotoxic metabolite doxorubicinol is increased in a mouse model of chronic glutathione deficiency: A potential role for carbonyl reductase 3. Chem.-Biol. Interact. 2015, 234, 154–161. [Google Scholar] [CrossRef] [Green Version]
  110. Yu, D.; Li, M.; Tian, Y.; Liu, J.; Shang, J. Luteolin inhibits ROS-activated MAPK pathway in myocardial ischemia/reperfusion injury. Life Sci. 2015, 122, 15–25. [Google Scholar] [CrossRef] [PubMed]
  111. Hu, W.; Xu, T.; Wu, P.; Pan, D.; Chen, J.; Chen, J.; Zhang, B.; Zhu, H.; Li, D. Luteolin improves cardiac dysfunction in heart failure rats by regulating sarcoplasmic reticulum Ca 2+-ATPase 2a. Sci. Rep. 2017, 7, 41017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Ding, X.; Zheng, L.; Yang, B.; Wang, X.; Ying, Y. Luteolin attenuates atherosclerosis via modulating signal transducer and activator of transcription 3-mediated inflammatory response. Drug Des. Dev. Ther. 2019, 13, 3899–3911. [Google Scholar] [CrossRef] [Green Version]
  113. Zhang, Y.; Ma, C.; Liu, C.; Wei, F. Luteolin attenuates doxorubicin-induced cardiotoxicity by modulating the PHLPP1/AKT/Bcl-2 signaling pathway. PeerJ 2020, 8, e8845. [Google Scholar] [CrossRef]
  114. Yao, H.; Shang, Z.; Wang, P.; Li, S.; Zhang, Q.; Tian, H.; Ren, D.; Han, X. Protection of luteolin-7-O-glucoside against doxorubicin-induced injury through PTEN/Akt and ERK pathway in H9c2 cells. Cardiovasc. Toxicol. 2016, 16, 101–110. [Google Scholar] [CrossRef] [PubMed]
  115. Gharanei, M.; Hussain, A.; Janneh, O.; Maddock, H.L. Doxorubicin-induced myocardial injury is exacerbated following ischaemic stress via an opening of the mitochondrial permeability transition pore. Toxicol. Appl. Pharmacol. 2013, 268, 149–156. [Google Scholar] [CrossRef] [PubMed]
  116. Wang, L.; Chen, Q.; Li, G.; Ke, D. Ghrelin stimulates angiogenesis via GHSR1a-dependent MEK/ERK and PI3K/Akt signal pathways in rat cardiac microvascular endothelial cells. Peptides 2012, 33, 92–100. [Google Scholar] [CrossRef]
  117. Xu, H.; Yu, W.; Sun, S.; Li, C.; Zhang, Y.; Ren, J. Luteolin attenuates doxorubicin-induced cardiotoxicity through promoting mitochondrial autophagy. Front. Physiol. 2020, 11, 113. [Google Scholar] [CrossRef] [PubMed]
  118. Wang, S.Q.; Han, X.Z.; Li, X.; Ren, D.M.; Wang, X.N.; Lou, H.X. Flavonoids from Dracocephalum tanguticum and their cardioprotective effects against doxorubicin-induced toxicity in H9c2 cells. Bioorg. Med. Chem. Lett. 2010, 20, 6411–6415. [Google Scholar] [CrossRef] [PubMed]
  119. Syahputra, R.A.; Harahap, U.; Dalimunthe, A.; Pandapotan, M.; Satria, D. Protective effect of Vernonia amygdalina Delile against doxorubicin-induced cardiotoxicity. Heliyon 2021, 7, e07434. [Google Scholar] [CrossRef] [PubMed]
  120. Azarabadi, S.; Abdollahi, H.; Torabi, M.; Salehi, Z.; Nasiri, J. ROS generation, oxidative burst and dynamic expression profiles of ROS-scavenging enzymes of superoxide dismutase (SOD), catalase (CAT), and ascorbate peroxidase (APX) in response to Erwinia amylovora in pear (Pyrus communis L). Eur. J. Plant Pathol. 2017, 147, 279–294. [Google Scholar] [CrossRef]
  121. Razavi-Azarkhiavi, K.; Iranshahy, M.; Sahebkar, A.; Shirani, K.; Karimi, G. The protective role of phenolic compounds against doxorubicin-induced cardiotoxicity: A comprehensive review. Nutr. Cancer 2016, 68, 892–917. [Google Scholar] [CrossRef] [PubMed]
  122. Hu, Y.; Zhang, C.; Zhu, H.; Wang, S.; Zhou, Y.; Zhao, J.; Xia, Y.; Li, D. Luteolin modulates SERCA2a via Sp1 upregulation to attenuate myocardial ischemia/reperfusion injury in mice. Sci. Rep. 2020, 10, 15407. [Google Scholar] [CrossRef]
  123. Kelly, G.S. Quercetin. Altern. Med. Rev. 2011, 16, 172–195. [Google Scholar]
  124. Sultana, B.; Anwar, F. Flavonols (kaempferol, quercetin, myricetin) contents of selected fruits, vegetables, and medicinal plants. Food Chem. 2008, 108, 879–884. [Google Scholar] [CrossRef]
  125. Batiha, G.E.; Beshbishy, A.M.; Ikram, M.; Mulla, Z.S.; El-Hack, M.E.; Taha, A.E.; Algammal, A.M.; Elewa, Y.H. The pharmacological activity, biochemical properties, and pharmacokinetics of the major natural polyphenolic flavonoid: Quercetin. Foods 2020, 9, 374. [Google Scholar] [CrossRef] [Green Version]
  126. Zhang, Y.M.; Zhang, Z.Y.; Wang, R.X. Protective mechanisms of quercetin against myocardial ischemia-reperfusion injury. Front. Physiol. 2020, 11, 956. [Google Scholar] [CrossRef] [PubMed]
  127. Chen, J.Y.; Hu, R.Y.; Chou, H.C. Quercetin-induced cardioprotection against doxorubicin cytotoxicity. J. Biomed. Sci. 2013, 20, 95. [Google Scholar] [CrossRef] [Green Version]
  128. Dong, Q.; Chen, L.; Lu, Q.; Sharma, S.; Li, L.; Morimoto, S.; Wang, G. Quercetin attenuates doxorubicin cardiotoxicity by modulating B mi-1 expression. Br. J. Pharmacol. 2014, 171, 4440–4454. [Google Scholar] [CrossRef] [Green Version]
  129. Chen, X.; Peng, X.; Luo, Y.; You, J.; Yin, D.; Xu, Q.; He, H.; He, M. Quercetin protects cardiomyocytes against doxorubicin-induced toxicity by suppressing oxidative stress and improving mitochondrial function via 14-3-3γ. Toxicol. Mech. Methods 2019, 29, 344–354. [Google Scholar] [CrossRef] [PubMed]
  130. Ahmed, Z.A.; Abtar, A.N.; Othman, H.H.; Aziz, T.A. Effects of quercetin, sitagliptin alone or in combination in testicular toxicity induced by doxorubicin in rats. Drug Des. Dev. Ther. 2019, 13, 3321–3329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Matouk, A.I.; Taye, A.; Heeba, G.H.; El-Moselhy, M.A. Quercetin augments the protective effect of losartan against chronic doxorubicin cardiotoxicity in rats. Environ. Toxicol. Pharmacol. 2013, 36, 443–450. [Google Scholar] [CrossRef]
  132. Al-Oanzi, Z.H.; Elasbali, A.M.; Alruwaili, N.K.; Alotaibi, N.H.; Alharbi, K.S.; Alzarea, A.I.; Alsuwayt, B.H.; Al-Enazi, M.M. Protective effect of baicalein alone and losartan-baicalein combination therapy on doxorubicin-induced hepatotoxicity in rats. Toxicol. Environ. Health Sci. 2020, 12, 45–54. [Google Scholar] [CrossRef]
  133. Sharma, A.; Parikh, M.; Shah, H.; Gandhi, T. Modulation of Nrf2 by quercetin in doxorubicin-treated rats. Heliyon 2020, 6, e03803. [Google Scholar] [CrossRef] [PubMed]
  134. Salehi, B.; Venditti, A.; Sharifi-Rad, M.; Kręgiel, D.; Sharifi-Rad, J.; Durazzo, A.; Lucarini, M.; Santini, A.; Souto, E.B.; Novellino, E.; et al. The therapeutic potential of apigenin. Int. J. Mol. Sci. 2019, 20, 1305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Ali, F.; Rahul; Naz, F.; Ali, F.; Jyoti, S.; Siddique, Y.H. Health functionality of apigenin: A review. Int. J. Food Prop. 2017, 20, 1197–1238. [Google Scholar] [CrossRef]
  136. Sahu, R.; Dua, T.K.; Das, S.; De Feo, V.; Dewanjee, S. Wheat phenolics suppress doxorubicin-induced cardiotoxicity via inhibition of oxidative stress, MAP kinase activation, NF-κB pathway, PI3K/Akt/mTOR impairment, and cardiac apoptosis. Food Chem. Toxicol. 2019, 125, 503–519. [Google Scholar] [CrossRef]
  137. Zare, M.F.; Rakhshan, K.; Aboutaleb, N.; Nikbakht, F.; Naderi, N.; Bakhshesh, M.; Azizi, Y. Apigenin attenuates doxorubicin-induced cardiotoxicity via reducing oxidative stress and apoptosis in male rats. Life Sci. 2019, 232, 116623. [Google Scholar] [CrossRef]
  138. Yu, W.; Sun, H.; Zha, W.; Cui, W.; Xu, L.; Min, Q.; Wu, J. Apigenin attenuates adriamycin-induced cardiomyocyte apoptosis via the PI3K/AKT/mTOR pathway. Evid.-Based Complement. Altern. Med. 2017, 2017, 2590676. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Quan, W.; Ma, S.; Zhu, Y.; Shao, Q.; Hou, J.; Li, X. Apigenin-7-O-β-d-(6″-p-coumaroyl)-glucopyranoside reduces myocardial ischaemia/reperfusion injury in an experimental model via regulating the inflammation response. Pharm. Biol. 2020, 58, 80–88. [Google Scholar] [CrossRef] [Green Version]
  140. Al-Dhabi, N.A.; Arasu, M.V.; Park, C.H.; Park, S.U. An up-to-date review of rutin and its biological and pharmacological activities. EXCLI J. 2015, 14, 59–63. [Google Scholar] [PubMed]
  141. Dagnon, S.; Novkova, Z.; Bojilov, D.; Nedialkov, P.; Kouassi, C. Development of surrogate standards approach for the determination of polyphenols in Vernonia amygdalina Del. J. Food Compos. Anal. 2019, 82, 103231. [Google Scholar] [CrossRef]
  142. Yang, C.-Y.; Hsiu, S.-L.; Wen, K.-C.; Lin, S.-P.; Tsai, S.-Y.; Hou, Y.-C.; Chao, P.-D. Bioavailability and metabolic pharmacokinetics of rutin and quercetin in rats. J. Food Drug Anal. 2005, 13, 5. [Google Scholar] [CrossRef]
  143. Panchal, S.K.; Poudyal, H.; Arumugam, T.V.; Brown, L. Rutin attenuates metabolic changes, nonalcoholic steatohepatitis, and cardiovascular remodeling in high-carbohydrate, high-fat diet-fed rats. J. Nutr. 2011, 141, 1062–1069. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Rodrigues, P.G.; Miranda-Silva, D.; Costa, S.M.; Barros, C.; Hamdani, N.; Moura, C.; Mendes, M.J.; Sousa-Mendes, C.; Trindade, F.; Fontoura, D.; et al. Early myocardial changes induced by doxorubicin in the nonfailing dilated ventricle. Am. J. Physiol.—Heart Circ. Physiol. 2019, 316, H459–H475. [Google Scholar] [CrossRef] [PubMed]
  145. Lee, K.H.; Cho, H.; Lee, S.; Woo, J.S.; Cho, B.H.; Kang, J.H.; Jeong, Y.M.; Cheng, X.W.; Kim, W. Enhanced-autophagy by exenatide mitigates doxorubicin-induced cardiotoxicity. Int. J. Cardiol. 2017, 232, 40–47. [Google Scholar] [CrossRef]
  146. Dirks-Naylor, A.J. The role of autophagy in doxorubicin-induced cardiotoxicity. Life Sci. 2013, 93, 913–916. [Google Scholar] [CrossRef] [PubMed]
  147. Guo, Z.; Tang, N.; Liu, F.Y.; Yang, Z.; Ma, S.Q.; An, P.; Wu, H.M.; Fan, D.; Tang, Q.Z. TLR9 deficiency alleviates doxorubicin-induced cardiotoxicity via the regulation of autophagy. J. Cell. Mol. Med. 1091, 24, 10913–10923. [Google Scholar] [CrossRef] [PubMed]
  148. Petroni, K.; Trinei, M.; Fornari, M.; Calvenzani, V.; Marinelli, A.; Micheli, L.A.; Pilu, R.; Matros, A.; Mock, H.P.; Tonelli, C.; et al. Dietary cyanidin 3-glucoside from purple corn ameliorates doxorubicin-induced cardiotoxicity in mice. Nutr. Metab. Cardiovasc. Dis. 2017, 27, 462–469. [Google Scholar] [CrossRef] [PubMed]
  149. Huang, P.C.; Kuo, W.W.; Shen, C.Y.; Chen, Y.F.; Lin, Y.M.; Ho, T.J.; Padma, V.V.; Lo, J.F.; Huang, C.Y. Anthocyanin attenuates doxorubicin-induced cardiotoxicity via estrogen receptor-α/β and stabilizes HSF1 to inhibit the IGF-IIR apoptotic pathway. Int. J. Mol. Sci. 2016, 17, 1588. [Google Scholar] [CrossRef] [Green Version]
  150. Lee, H.J.; Lee, W.J.; Chang, S.E.; Lee, G.Y. Hesperidin, a popular antioxidant inhibits melanogenesis via Erk1/2 mediated MITF degradation. Int. J. Mol. Sci. 2015, 16, 18384–18395. [Google Scholar] [CrossRef] [Green Version]
  151. Trivedi, P.P.; Kushwaha, S.; Tripathi, D.N.; Jena, G.B. Cardioprotective effects of hesperetin against doxorubicin-induced oxidative stress and DNA damage in rats. Cardiovasc. Toxicol. 2011, 11, 215–225. [Google Scholar] [CrossRef]
  152. Saad, S.; Ahmad, I.; Kawish, S.M.; Khan, U.A.; Ahmad, F.J.; Ali, A.; Jain, G.K. Improved cardioprotective effects of hesperidin solid lipid nanoparticles prepared by supercritical antisolvent technology. Colloids Surf. B Biointerfaces 2020, 187, 110628. [Google Scholar] [CrossRef] [PubMed]
  153. Doerr, V.; Montalvo, R.N.; Kwon, O.S.; Talbert, E.E.; Hain, B.A.; Houston, F.E.; Smuder, A.J. Prevention of doxorubicin-induced autophagy attenuates oxidative stress and skeletal muscle dysfunction. Antioxidants 2020, 9, 263. [Google Scholar] [CrossRef] [Green Version]
  154. Mani, R.; Natesan, V. Chrysin: Sources, beneficial pharmacological activities, and molecular mechanism of action. Phytochemistry 2018, 145, 187–196. [Google Scholar] [CrossRef]
  155. Mantawy, E.M.; Esmat, A.; El-Bakly, W.M.; ElDin, R.A.; El-Demerdash, E. Mechanistic clues to the protective effect of chrysin against doxorubicin-induced cardiomyopathy: Plausible roles of p53, MAPK and AKT pathways. Sci. Rep. 2017, 7, 4795. [Google Scholar] [CrossRef] [Green Version]
  156. Mantawy, E.M.; El-Bakly, W.M.; Esmat, A.; Badr, A.M.; El-Demerdash, E. Chrysin alleviates acute doxorubicin cardiotoxicity in rats via suppression of oxidative stress, inflammation and apoptosis. Eur. J. Pharmacol. 2014, 728, 107–118. [Google Scholar] [CrossRef] [PubMed]
  157. Salehi, B.; Fokou, P.V.; Sharifi-Rad, M.; Zucca, P.; Pezzani, R.; Martins, N.; Sharifi-Rad, J. The therapeutic potential of naringenin: A review of clinical trials. Pharmaceuticals 2019, 12, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Chlebowski, R.T. Adriamycin (doxorubicin) cardiotoxicity: A review. West. J. Med. 1979, 131, 364. [Google Scholar]
  159. Shiromwar, S.S.; Chidrawar, V.R. Combined effects of p-coumaric acid and naringenin against doxorubicin-induced cardiotoxicity in rats. Pharmacogn. Res. 2011, 3, 214–219. [Google Scholar] [CrossRef] [Green Version]
  160. Abd-Allah, A.R.; Al-Majed, A.A.; Mostafa, A.M.; Al-Shabanah, O.A.; Din, A.G.E.; Nagi, M.N. Protective effect of arabic gum against cardiotoxicity induced by doxorubicin in mice: A possible mechanism of protection. J. Biochem. Mol. Toxicol. 2002, 16, 254–259. [Google Scholar] [CrossRef]
  161. Kwatra, M.; Kumar, V.; Jangra, A.; Mishra, M.; Ahmed, S.; Ghosh, P.; Vohora, D.; Khanam, R. Ameliorative effect of naringin against doxorubicin-induced acute cardiac toxicity in rats. Pharm. Biol. 2016, 54, 637–647. [Google Scholar] [CrossRef] [PubMed]
  162. Subburaman, S.; Ganesan, K.; Ramachandran, M. Protective role of naringenin against doxorubicin-induced cardiotoxicity in a rat model: Histopathology and mRNA expression profile studies. J. Environ. Pathol. Toxicol. Oncol. 2014, 33, 363–376. [Google Scholar] [CrossRef]
  163. Kaiserová, H.; Šimůnek, T.; van der Vijgh, W.J.; Bast, A.; Kvasničková, E. Flavonoids as protectors against doxorubicin cardiotoxicity: Role of iron chelation, antioxidant activity and inhibition of carbonyl reductase. Biochim. Biophys. Acta—Mol. Basis Dis. 2007, 1772, 1065–1074. [Google Scholar] [CrossRef] [Green Version]
  164. Cote, B.; Carlson, L.J.; Rao, D.A.; Alani, A.W. Combinatorial resveratrol and quercetin polymeric micelles mitigate doxorubicin-induced cardiotoxicity in vitro and in vivo. J. Control. Release 2015, 213, 128–133. [Google Scholar] [CrossRef] [PubMed]
  165. Wang, Y.; Zhang, Y.; Sun, B.; Tong, Q.; Ren, L. Rutin protects against pirarubicin-induced cardiotoxicity through the TGF-β1-p38 MAPK signaling pathway. Evid.-Based Complement. Altern. Med. 2017, 2017, 1759385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Ma, Y.; Yang, L.; Ma, J.; Lu, L.; Wang, X.; Ren, J.; Yang, J. Rutin attenuates doxorubicin-induced cardiotoxicity via regulating autophagy and apoptosis. Biochim. Biophys. Acta—Mol. Basis Dis. 2017, 1863, 1904–1911. [Google Scholar] [CrossRef] [PubMed]
  167. Hozayen, W.G.; Abou Seif, H.S. Protective effects of rutin and hesperidin against doxorubicin-induced lipodystrophy and cardiotoxicity in albino rats. J. Am. Sci. 2011, 7, 765–775. [Google Scholar]
  168. Tang, S.; Kan, J.; Sun, R.; Cai, H.; Hong, J.; Jin, C.; Zong, S. Anthocyanins from purple sweet potato alleviate doxorubicin-induced cardiotoxicity in vitro and in vivo. J. Food Biochem. 2021, 45, e13869. [Google Scholar] [CrossRef] [PubMed]
  169. Donia, T.I.; Gerges, M.N.; Mohamed, T.M. Amelioration effect of Egyptian sweet orange hesperidin on Ehrlich ascites carcinoma (EAC) bearing mice. Chem.-Biol. Interact. 2018, 285, 76–84. [Google Scholar] [CrossRef] [PubMed]
  170. Arafa, H.M.; Abd-Ellah, M.F.; Hafez, H.F. Abatement by naringenin of doxorubicin-induced cardiac toxicity in rats. J. Egypt. Natl. Cancer Inst. 2005, 17, 291–300. [Google Scholar]
  171. Han, X.; Gao, S.; Cheng, Y.; Sun, Y.; Liu, W.; Tang, L.; Ren, D. Protective effect of naringenin-7-O-glucoside against oxidative stress induced by doxorubicin in H9c2 cardiomyocytes. Biosci. Trends 2012, 6, 19–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Doxorubicin mechanism of cardiotoxicity is doxorubicin converted into doxorubicin-semiquinone by transferring an electron from NADPH, while it changes into NADP+ continuously. However, an electron is transferred from O2 to doxorubicin-semiquinone, thereby creating O2 (superoxide radical) neutralized by SOD into H2O2 which can be converted into H2O + O2. In cardiac toxicity events, the SOD and CAT leaves are down; hence, H2O2 is converted into OH and *OOH (hydroperoxyl radical) by the Fenton reaction. The superoxide radical is highly active, such that it directly damages the cell membrane, specifically the mitochondria membrane, which causes the increase in lipid peroxidation, and the membrane permeability of mitochondria also causes ATP loss. On the other hand, the superoxide radical also triggers the stimulation of protein P38 and JNK which activates protein p53 and increases caspase 3 activity. Furthermore, cytochrome c was released and activated the Bax (pro-apoptosis protein) which stimulates the activation of caspase 3 activity which increases apoptosis events. The Dox mechanism of action binding into topoisomerase 2β breaks the DNA that causes apoptosis of the cell. Additionally, ROS directly damages the reticulum sarcoplasm that causes the elevation of Ca2+ into the cytosol and increases the contractile, thereby causing contractile impairment. The accumulation of apoptosis in cardiac cells and contractile impairment leads to cardiomyopathy and the release of cardiac biomarkers such as Troponin T, CK-MB, LDH, BNP, NT-pro-BNP, ANP, and CRP.
Figure 1. Doxorubicin mechanism of cardiotoxicity is doxorubicin converted into doxorubicin-semiquinone by transferring an electron from NADPH, while it changes into NADP+ continuously. However, an electron is transferred from O2 to doxorubicin-semiquinone, thereby creating O2 (superoxide radical) neutralized by SOD into H2O2 which can be converted into H2O + O2. In cardiac toxicity events, the SOD and CAT leaves are down; hence, H2O2 is converted into OH and *OOH (hydroperoxyl radical) by the Fenton reaction. The superoxide radical is highly active, such that it directly damages the cell membrane, specifically the mitochondria membrane, which causes the increase in lipid peroxidation, and the membrane permeability of mitochondria also causes ATP loss. On the other hand, the superoxide radical also triggers the stimulation of protein P38 and JNK which activates protein p53 and increases caspase 3 activity. Furthermore, cytochrome c was released and activated the Bax (pro-apoptosis protein) which stimulates the activation of caspase 3 activity which increases apoptosis events. The Dox mechanism of action binding into topoisomerase 2β breaks the DNA that causes apoptosis of the cell. Additionally, ROS directly damages the reticulum sarcoplasm that causes the elevation of Ca2+ into the cytosol and increases the contractile, thereby causing contractile impairment. The accumulation of apoptosis in cardiac cells and contractile impairment leads to cardiomyopathy and the release of cardiac biomarkers such as Troponin T, CK-MB, LDH, BNP, NT-pro-BNP, ANP, and CRP.
Molecules 27 01320 g001
Figure 2. Flavonoid subclass structures (the same color represents the same subclasses of flavonoids).
Figure 2. Flavonoid subclass structures (the same color represents the same subclasses of flavonoids).
Molecules 27 01320 g002
Figure 3. Summary of flavonoids’ role against doxorubicin that increases (green) antioxidant endogen, cardiac function, mitochondria function, calcium homeostasis, nerf 2 expressions, and ATP while reducing (red) inflammatory, ROS, apoptosis, lipid peroxidation, caspase 3 activity.
Figure 3. Summary of flavonoids’ role against doxorubicin that increases (green) antioxidant endogen, cardiac function, mitochondria function, calcium homeostasis, nerf 2 expressions, and ATP while reducing (red) inflammatory, ROS, apoptosis, lipid peroxidation, caspase 3 activity.
Molecules 27 01320 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Syahputra, R.A.; Harahap, U.; Dalimunthe, A.; Nasution, M.P.; Satria, D. The Role of Flavonoids as a Cardioprotective Strategy against Doxorubicin-Induced Cardiotoxicity: A Review. Molecules 2022, 27, 1320. https://doi.org/10.3390/molecules27041320

AMA Style

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. https://doi.org/10.3390/molecules27041320

Chicago/Turabian Style

Syahputra, Rony Abdi, Urip Harahap, Aminah Dalimunthe, M. Pandapotan Nasution, and Denny Satria. 2022. "The Role of Flavonoids as a Cardioprotective Strategy against Doxorubicin-Induced Cardiotoxicity: A Review" Molecules 27, no. 4: 1320. https://doi.org/10.3390/molecules27041320

Article Metrics

Back to TopTop