Next Article in Journal
Paeoniflorin Inhibits the Activation of Microglia and Alleviates Depressive Behavior by Regulating SIRT1-NF-kB-NLRP3/Pyroptosis Pathway
Previous Article in Journal
A Recombinant Shigella flexneri Strain Expressing ETEC Heat-Labile Enterotoxin B Subunit Shows Promise for Vaccine Development via OMVs
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Syzygium aromaticum Extract Mitigates Doxorubicin-Induced Hepatotoxicity in Male Rats

by
Alaa Muqbil Alsirhani
1,
Amal S. Abu-Almakarem
2,
Maha Abdullah Alwaili
3,
Salwa Aljohani
4,
Ibtisam Alali
1,
Aljazi Abdullah AlRashidi
5,
Najlaa Yousef Abuzinadah
6,
Sahar Abdulrahman Alkhodair
7,
Maysa A. Mobasher
8,
Tahiyat Alothaim
9,
Thamir M. Eid
7 and
Karim Samy El-Said
10,*
1
Department of Chemistry, College of Science, Jouf University, Sakaka 72341, Saudi Arabia
2
Department of Basic Medical Sciences, Faculty of Applied Medical Sciences, Al-Baha University, Al Baha 65431, Saudi Arabia
3
Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia
4
Chemistry Department, Faculty of Science, Taibah University, Yanbu Branch, Yanbu 46423, Saudi Arabia
5
Chemistry Department, Faculty of Science, University of Ha’il, Ha’il 81451, Saudi Arabia
6
Department of Biological Science, College of Science, University of Jeddah, Jeddah 23714, Saudi Arabia
7
Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
8
Department of Pathology, Biochemistry Division, College of Medicine, Jouf University, Sakaka 72388, Saudi Arabia
9
Department of Biology, College of Science, Qassim University, Buraydah 51452, Saudi Arabia
10
Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta 31527, Egypt
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(23), 12541; https://doi.org/10.3390/ijms252312541
Submission received: 7 October 2024 / Revised: 9 November 2024 / Accepted: 19 November 2024 / Published: 22 November 2024
(This article belongs to the Section Biochemistry)

Abstract

:
Doxorubicin (DOX), an anticancer drug, is used to treat several types of tumors, but it has detrimental side effects that restrict its therapeutic efficacy. One is the iron-dependent form of ferroptosis, which is characterized by elevated ROS production and iron overload. Syzygium aromaticum has a diverse range of biological and pharmaceutical actions due to their antioxidant properties. This study investigated the effect of S. aromaticum extract (SAE) on hepatotoxicity caused by DOX in rats. Phytochemical analysis was performed to assess compounds in SAE. The ADMETlab 2.0 web server was used to predict the pharmacokinetic properties of the most active components of SAE when DOX was injected into rats. Molecular docking studies were performed using AutoDock Vina. Forty male Sprague Dawley rats were divided into four groups of ten rats each (G1 was a negative control group, G2 was given 1/10 of SAE LD50 by oral gavage (340 mg/kg), G3 was given 4 mg/kg of DOX intraperitoneally (i.p.) once a week for a month, and G4 was administered DOX as in G3 and SAE as in G2). After a month, biochemical and histopathological investigations were performed. Rats given SAE had promising levels of phytochemicals, which could significantly ameliorate DOX-induced hepatotoxicity by restoring biochemical alterations, mitigating ferroptosis, and upregulating the NRF-2–SLC7A-11–GPX-4 signaling pathway. These findings suggest that SAE could potentially alleviate DOX-induced hepatotoxicity in rats.

1. Introduction

Doxorubicin (DOX) is a chemotherapeutic anthracycline that is used to treat various types of cancer; however, it has some side effects that limit its efficacy, such as encouraging the accumulation of hazardous intermediates that have been linked to the production of liver injury [1]. Several studies have reported that DOX treatment negatively affected liver tissues and was associated with distinct biochemical and histopathological alterations [2,3]. Oxidative stress, apoptosis, and inflammation in the liver tissues were the main mechanisms with which DOX induced hepatotoxicity [4]. Ferroptosis is one of the recently identified molecular pathways associated with the pathophysiology of several disorders [5]. It is a major factor in the development of liver injury and is a possible pharmaceutical target [6]. The main biochemical features of ferroptosis include intracellular iron overload, lipid peroxidation, and antioxidant imbalance [7].
Nuclear factor E2-related factor 2 (NRF-2) regulates the core antioxidant response that controls essential defensive mechanisms in cells and protects against cellular damage [8]. A recent investigation reported that the regulation of solute carrier family 7, member 11 (SLC7A-11) had a role in protecting cells against ferroptotic death, and that this aided in the production of glutathione (GSH) [9]. Other proteins that are closely linked to ferroptosis are ferritin heavy polypeptide 1 (FTH-1) and glutathione peroxidase 4 (GPX-4) [10]. A previous study demonstrated the molecular mechanisms and therapeutic targeting of ferroptosis in DOX-induced organ toxicity [11]. Furthermore, it has been reported that regulating the NRF2–GPx4 axis with natural compounds inhibits ferroptosis in CCl4-induced liver injury in mice [12]. Ferritinophagy was mediated by DOX, which releases significant amounts of redox-active iron. The biochemical process leading to ferroptosis is Fenton’s reaction, which causes an excessive build up of ferrous iron (Fe2+) in the cells [13]. Therefore, the pharmacological targeting of ferroptosis could be a novel interventional modality for inhibiting DOX-induced toxicity in patients receiving DOX chemotherapy.
Natural antioxidants connect to ferrous iron (Fe2+), thereby preventing the generation of reactive intermediates that harm cells [14,15]. There have been reports on the preventive and therapeutic uses of herbal constituents that act against DOX-induced hepatotoxicity [16]. A previous report indicated a beneficial role of naringin in preventing DOX-induced liver damage; it did this by upregulating sirtuin 1 (SIRT1), which lowered oxidative stress, inflammation, and apoptosis [17]. Another study reported on the potential ameliorative effects of beetroot ethanolic extract against DOX-induced hepatic damage in rats [18]. Allium cepa extract indicated against liver damage caused by DOX in rats [19]. Syzygium aromaticum (clove), belonging to the Myrtaceae family, is cultivated in Brazil, Egypt, Madagascar, and Morocco [20]. It has several biological, biomedical, gastronomic, and traditional uses in medicine because of its high concentration of bioactive ingredients, including eugenol and flavonoids [21]. Some of its numerous pharmaceutical activities include antimicrobial, antiparasitic, antioxidant, anti-inflammatory, and anesthetic effects [22]. Pourlak et al. (2020) reported that S. aromaticum extract (SAE) treatment led to a significant mitigation of hepatic cell damage and oxidative stress in rats [23]. Furthermore, SAE significantly reduced levels of liver function enzymes and inflammatory cytokines in rats intoxicated with CCl4 [24]. It has been reported that SAE markedly improved the antioxidant status of Wistar rats intoxicated with artesunate [25]. The ameliorative efficacy of SAE has been reported to be associated with behavioral changes in lead-induced neurotoxicity in mice [26]. This novel study investigated the efficacy of SAE in ameliorating DOX-promoted liver damage in male rats.

2. Results

2.1. Phytochemical Components and GC-MS Analysis of SAE

The results show that the S. aromaticum flower buds (SAFBs) yielded an adequate extract amount (14%). The total phenolic and flavonoid contents of ASF were represented by 31.86 ± 2.43 mg GAE/g DW and 18.65 ± 1.58 mg QE/g DW, respectively. The TAC of SAE was 69.37 ± 3.79 mg AAE/g DW. The saponin and anthocyanin levels were 365 ± 4.37 mg/g DW and 7.89 ± 0.45 mg ECG/g DW, respectively. The DPPH scavenging activity was 82.64% ± 3.95. The IC50 was 6.05 ± 0.86 mg/mL (Table 1).
GC-MS analysis showed that SAE contains promising phytochemicals, the most abundant being ethyl-α-D-glucopyranoside, eugenol, caryophyllene, and humulene. The retention times (RTs) were 7.64, 12.25, 15.82, and 16.54 min, respectively. The peak area percentages (PA%) were 2.27%, 64.17%, 18.07%, and 3.83%, respectively (Figure 1 and Table 2).

2.2. In Silico ADMET Analysis

ADMET screening provided a comprehensive comparison of the pharmacokinetic and toxicological properties of DOX and revealed that eugenol was the most abundant compound in SAE. Eugenol had better oral bioavailability (F20% and F30%) and blood–brain barrier penetration (BBB) than DOX. DOX has a high likelihood of being a P-glycoprotein substrate, which may affect its cellular accumulation and efficacy. Furthermore, eugenol and 10-Heptadecen-8-ynoic acid, methyl ester showed plasma-protein-binding affinities with lower fractions unbound (Fu) than DOX, indicating its active transport through the bloodstream. In addition, eugenol demonstrated a lower probability of various toxic endpoints. The toxicity prediction results for eugenol and DOX further highlight the differences in their safety profiles. Eugenol and 10-Heptadecen-8-ynoic acid, methyl ester are predicted to be inactive against most organ toxicities. In contrast, DOX was predicted to be active versus multiple organ toxicities, including cardiotoxicity and hepatotoxicity (Figure 2 and Table 3). The bioconcentration factors (BCFs) of eugenol and 10-Heptadecen-8-ynoic acid (1.203) and methyl ester (2.424) were higher than those of DOX (0.567), which suggests their high degree of uptake from the dissolved phase. The quantitative estimate of drug-likeness (QED) showed that eugenol and 10-Heptadecen-8-ynoic acid, methyl ester were closer to 1, indicating that they were more drug-like than DOX. The Lipinski filter is used to filter out any drug at the absorption or permeation level (an ideal drug has a molecular weight of less than 500 g/mol). The molecular weight of eugenol was 164.08 g/mol and that of 10-Heptadecen-8-ynoic acid, methyl ester was 278.22 g/mol, so they were acceptable, while that of DOX (543.17 g/mol) was not. The Veber (GSK) rule defines drug-likeness constraints as a rotatable bond count ≤ 10 and a topological polar surface area (TPSA) ≤ 140. Our ADMET screening showed that eugenol and 10-Heptadecen-8-ynoic acid, methyl ester followed the rule and were acceptable by GSK standards, while DOX was not (Table 3). The AMES test indicated that toxicity, hepatotoxicity, and skin sensitization were not found in the therapeutic compounds compared with DOX. The overall analysis demonstrated that eugenol was more biodegradable and bioavailable and has physicochemical, molecular, and ADMET properties that lie between the upper and lower predicted values (see Figure 2 and Table 3).

2.3. Bioactive Compound (Eugenol) Interactions with Target Proteins (NRF-2, SLC7A-11, and GPX-4) as Shown by Molecular Docking Analysis

The molecular docking results presented in Table 4 show the ∆G and binding affinities of the compounds detected in SAE and DOX for the key proteins involved in cellular protection and the oxidative stress response (NRF-2, SLC7A-11, and GPX-4). DOX consistently demonstrated stronger binding affinities across the three targets NRF-2 (−7.8), SLC7A-11 (−8.4), and GPX-4 (−6.7) compared with other compounds. 10-heptadecen-8-ynoic acid, methyl ester and eugenol showed lower binding affinities (Table 4). This suggests that DOX may have a greater inhibitory effect on these proteins, and this could contribute to its potential toxicity. DOX formed more hydrogen bonds and had a greater variety of interactions than 10-heptadecen-8-ynoic acid, methyl ester (NRF-2 (−4.3), SLC7A-11 (−5.3), and GPX-4 (−3.9)) and eugenol (NRF-2 (−4.7), SLC7A-11 (−6.0), and GPX-4 (−4.3)). This extensive binding pattern of DOX could potentially disrupt GPX-4 function significantly, which might compromise the cells’ antioxidant defenses. The interactions with NRF-2 showed a similar trend, with DOX forming multiple hydrogen bonds and hydrophobic interactions, whereas eugenol primarily engaged in hydrophobic interactions. NRF-2 is a crucial regulator of the cellular response to oxidative stress, and the stronger binding of DOX might interfere with its protective functions to a greater extent than eugenol. For SLC7A-11, both compounds showed fewer interactions than the other targets, but DOX still demonstrated stronger binding. SLC7A-11 is involved in glutathione synthesis, an important cellular antioxidant. The different binding patterns suggest that DOX might have a greater impact on this cytoprotective pathway (Figure 3, Figure 4 and Figure 5).

2.4. The Oral LD50 of SAE, Body Weight, and Liver Weight Changes After Treatment with DOX–SAE

The LD50 of SAE after oral administration was determined for dosages of 1000 to 5000 mg/kg in different groups of rats after 24 h of treatment. The rats showed no stereotypical toxic symptoms except at 3400 mg/kg. The probit analysis showed that the oral LD50 of SAE was 3400 mg/kg.
The DOX-injected group showed a significant decrease (p < 0.05) in % b. wt. change (16.42%) compared with that in the control groups. Treatment of DOX-exposed rats with SAE led to a significant improvement (p < 0.05) in % b. wt. changes to 28.75% compared with that in the DOX-treated group (Figure 6A). Among the experimental groups under the study, the relative liver weight of the DOX-injected group increased (see Figure 6B).

2.5. Treatment with SAE Restored Hepatic Function Markers in DOX-Injected Rats

In the DOX-injected group, levels of liver transaminases (ALT and AST) were significantly increased (p < 0.05) (to 71.23 ± 1.78 and 103.67 ± 2.78 U/L, respectively). However, the treatment of DOX-injected rats with SAE led to a considerable reduction in the ALT and AST levels (to 38.93 ± 0.85 and 60.78 ± 2.56 U/L, respectively) (Table 5). Furthermore, the results show that DOX-exposed rats showed a significant increase (p < 0.05) in ALP level, representing 486.98 ± 6.34 U/L compared with the control groups. However, treatment with SAE demonstrated a significant decrease in ALP serum level (317.92 ± 5.79 U/L) compared with the DOX-injected group alone. The total and direct bilirubin levels in the DOX-injected group increased significantly (p < 0.05), but SAE therapy decreased these levels (see Table 5).

2.6. Treatment with SAE Alleviated Hepatic Oxidative Stress Induced by DOX in Rats

The DOX-treated rats showed a significant increase (p < 0.05) in their hepatic MDA levels (to 0.542 ± 0.025 nmol/mg protein) versus the negative control (0.258 ± 0.019 nmol/mg protein) and SAE control groups (0.231 ± 0.014 nmol/mg protein) (Figure 7A). Hepatic SOD and CAT activities significantly decreased (p < 0.05) in the hepatic tissues of DOX-injected rats when compared with the control groups; however, concomitant treatment with DOX and SAE led to a significant restoration of SOD and CAT activities (Figure 7B,C). Moreover, the GSH level was significantly decreased in the DOX-injected group (by −2.2 fold) compared with the control groups (Figure 7B). The group that was treated with DOX–SAE showed a significant increase in the GSH level as compared with the DOX-administered group alone (25.15 ± 1.14 vs. 14.12 ± 0.48 mg/mg protein) (Figure 7D).

2.7. SAE Treatment Modulated Ferroptosis-Related Proteins and Their Gene Expressions in DOX-Exposed Rats

Figure 8A demonstrates that, compared with the hepatic iron level in the negative control and SAE control groups (2.1 ± 0.16 and 1.8 ± 0.14 µmol/g protein, respectively), the DOX-injected group had a significant increase in hepatic iron levels (4.72 ± 0.31, p < 0.01). The group treated with DOX–SAE had a significant reduction (p < 0.01) in the hepatic iron level. To investigate the effect of DOX–SAE treatment on specific ferroptosis-related parameters in rats, the levels of NRF-2, SLC7A-11, and GPX-4 were determined in the rats’ livers. There were significant reductions (p < 0.01) in these proteins in the DOX-treated group compared with those in the control groups. However, treatment with SAE led to a significant restoration of the previously mentioned ferroptosis-related parameters in hepatic rats when compared with the DOX-injected group alone (see Figure 8).
The total protein levels in the hepatic tissues were measured using the method of Lowry et al. (1951) with bovine serum albumin as a standard. The DOX-exposed group showed a significant decrease (p < 0.05) in the hepatic protein concentration, whereas the combination of DOX and SAE treatments resulted in a significant increase in hepatic protein concentrations. Furthermore, compared with the negative control and SAE control groups (28.2 ± 0.98 and 31.1 ± 1.10 ng/mg protein, respectively), the protein level of FTH-1 in the DOX-injected group was considerably lower in the rats’ hepatocytes (13.3 ± 0.79 ng/mg protein, p < 0.05). Meanwhile, the DOX–SAE-treated group showed a significant increase in the FTH-1 level (21.2 ± 0.95 ng/mg protein, p < 0.05) compared with the DOX-injected group alone. In contrast, ACSL-4 and NCOA-4 protein levels were significantly increased in the DOX-treated group versus the control groups. However, these increases in ACSL-4 and NCOA-4 levels were significantly attenuated (p < 0.05) in the setting of DOX–SAE treatment (Figure 9).
Molecular analysis of ferroptosis-related genes by the RT-PCR method using glyceraldehyde-3-phosphate dehydrogenase (GAPDH) as a housekeeping gene showed that DOX injection in rats led to a significant downregulation (p < 0.001) in the relative mRNA expression levels of the NRF2, SLC7A11, GPX4, and FTH1 genes (by 2.5, 1.6, 1.4, and 4-fold, respectively). In contrast, the fold changes in the NCOA4 gene were significantly upregulated (p < 0.001) in the DOX-challenged group (by 3.7 fold) (Table 6). Interestingly, concomitant treatment with DOX and SAE led to a significant upregulation of NRF2, SLC7A11, GPX4, and FTH1 along with a significant downregulation of NCOA4 (see Table 6).

2.8. Treatment with SAE Reduced Hepatic Inflammatory Cytokines in Rats Given DOX

The results show that, in the group injected with DOX alone, the levels of inflammatory biomarkers, including IL-6, IL-1β, TNF-α, NF-κB, and COX-2, were significantly elevated (p < 0.001) compared with the control groups. However, these inflammatory cytokines were significantly decreased (p < 0.01) in the DOX–SAE-administered group (Table 7).

2.9. Treatment with SAE Improved Hepatic Histopathological Changes Induced by DOX in Rats

In H&E-stained liver sections, histopathological analysis revealed a normal hepatocyte architecture and a central hepatic vein with centrally located nuclei in the liver sections of the negative control group and the group given SAE; their pathological scores were 0.12 ± 0.09 and 0.10 ± 0.11, respectively (Table 8, Figure 10A,B). The liver sections of DOX-treated rats showed extensive hepatocyte degeneration, with an acutely dilated central vein, cellular swelling, and nuclear changes. The semi-quantitative analysis showed high pathological scores (3.50 ± 0.21; Table 8, Figure 10C). The liver section of the group given DOX–SAE displayed a significant improvement in the hepatic architecture and less congestion (see Table 8 and Figure 10D).

3. Discussion

The anthracycline DOX is one of the most commonly used systemic anti-neoplastic drugs. Its clinical efficacy is hampered, however, by the toxicities it induces. Hepatotoxicity is a frequent, serious adverse consequence caused by oxidative stress [2]. To date, there have been no specific and efficient treatment options available for the hepatotoxicity promoted by DOX. Therefore, it is imperative to evaluate natural constituents that might raise the therapeutic index of DOX while lowering its adverse effects [4]. Various natural constituents have been reported to exert certain curative effects on liver injuries by targeting ferroptosis [27]. Clove is one of the spices used in various dishes and is interestingly utilized for several therapeutic uses in traditional medicine related to its bioactive chemical constituents [28]. NRE-2 plays a key role in the process of ferroptosis by regulating a series of proteins, including FTH-1, GPX-4, SLC7A-11, and HO-1 [29]. Interestingly, some plant extracts could be effectively used as therapeutic targets to alleviate liver injuries by regulating NRF-2 or downstream effector proteins inhibiting ferroptosis [5,30]. Recently, a noted increase in the expression of GPX-4 and SLC7A-11 in liver tissues suggested that natural products may inhibit ferroptosis [31]. The current investigation addressed the ameliorative effect of SAE treatment on DOX-induced hepatotoxicity in male rats.
The results obtained from the current study show that, as demonstrated in Table 1, the flower buds of S. aromaticum yielded an adequate amount of extract and contained promising phytochemical constituents, including phenols, flavonoids, saponins, and anthocyanins, and have high percentages of DPPH scavenging activity [20,21,28,32]. Furthermore, GC-MS analysis of SAE revealed the presence of pharmacologically important phytochemical compounds, including ethyl-α-D-glucopyranoside, eugenol, caryophyllene, and humulene (see Table 2 and Figure 1). Consistently with our investigation, previous reports demonstrated that SAE contains bioactive compounds and that eugenol was the richest compound found when different extraction methods were used [28,33].
In the context of cytoprotection and cytotoxicity in hepatic cells, the present results suggest that eugenols may offer a more favorable safety profile, while the stronger interactions of DOX with antioxidants and stress-response proteins might contribute to its hepatotoxic effects [4]. By examining the types of interactions and their distances, we can infer the strength and specificity of binding, which can correlate with biological activity. For GPX-4, DOX forms multiple conventional hydrogen bonds with distances of 2.00 to 2.81 Å. These short hydrogen bonds indicate strong, specific interactions that are likely to significantly influence the functioning of GPX-4 [34]. Additionally, DOX was engaged in electrostatic and hydrophobic interactions, contributing to a stable binding pose. In contrast, eugenol’s interaction with GPX-4 was characterized by a single conventional hydrogen bond (2.24 Å) and several hydrophobic interactions. The fewer and generally weaker interactions suggest that eugenol may have had a more moderate effect on GPX-4 activity than DOX. With NRF-2, DOX demonstrated multiple conventional hydrogen bonds with distances between 2.05 and 2.55 Å, indicating strong binding. The presence of several hydrophobic interactions further stabilizes the binding [35]. Eugenol’s interaction with NRF-2 is limited to hydrophobic interactions, with no hydrogen bonds observed. This suggests a weaker and potentially less specific binding to NRF-2 compared with DOX, which may have led to a reduced impact on NRF2-mediated pathways [36]. However, for SLC7A-11, eugenol shows two conventional hydrogen bonds with distances of 2.13 and 2.97 Å, along with hydrophobic interactions. This binding pattern suggests a moderate interaction with SLC7A-11. The interaction of DOX with SLC7A-11 is characterized by a single conventional hydrogen bond (2.07 Å) and two pi–alkyl interactions. The fewer interactions observed for both compounds with SLC7A-11 than the other targets may indicate that this protein is less significantly affected by either compound. Specifically, the conventional hydrogen bonds, particularly those under 3 Å, were crucial for specific and strong binding [37]. DOX consistently formed more of these bonds across all targets, suggesting it may more potently modulate the activities of these proteins. Therefore, hydrophobic interactions played a role in the binding stability. While these interactions were generally weaker than hydrogen bonds, their cumulative effect could significantly impact the binding affinity. DOX formed some electrostatic interactions, particularly with GPX-4, which could enhance the binding specificity and strength. In addition, the interaction distances for the shorter interaction distances observed (mostly between 2 and 4 Å) indicate strong binding for both compounds, with DOX generally showing shorter distances and potentially stronger interactions. The diversity of the interactions, especially DOX, consistently demonstrates a more diverse range of interaction types across all targets, suggesting a more complex and potentially more disruptive binding mode [38]. These interaction patterns reveal that, while both compounds could bind to these cytoprotective proteins, DOX’s bindings are generally stronger and more extensive. This could explain its higher potency and its greater potential for disrupting normal cellular functions, which could lead to side effects [39].
At a dose of 1000 mg/kg body weight per day, cloves have been reported to have no toxic effects on Wistar rats [40]. The present study found that the oral LD50 of SAE was 3400 mg/kg after 24 h of treatment in rats. A previous study on oral toxicity revealed that the LD50 of S. aromaticum essential oil was approximately 4500 mg/kg, demonstrating the safety of SAE oral administration [41]. Treatment with DOX–SAE led to a significant improvement in the % b. wt. and relative liver weight changes (Figure 6). This finding was consistent with previous research showing the impact of natural plant constituents on DOX-induced body weight loss in experimental animals [4,42]. Our findings demonstrate that SAE treatment could reduce elevated ALT, AST, and ALP activities as well as the total and direct bilirubin levels compared with those activities in DOX-injected rats, indicating the therapeutic properties of SAE against DOX-promoted liver damage. These results are in line with those of numerous studies highlighting the hepatoprotective potential of plant extracts to limit DOX-induced liver injuries in experimental animals [4,24,43,44]. Additionally, data from our study reveal that DOX injection in rats resulted in a significant increase in their hepatic MDA levels and a significant decrease in their hepatic SOD, CAT, and GSH levels (see Figure 7). This could be attributed to the increased production of reactive free radicals by the DOX-exposed liver tissues, and, hence, the peroxidation of the lipid membrane and increased MDA generation [4,45]. Increased lipid peroxidation damages liver tissue and impairs antioxidant defense mechanisms, increasing the risk that metabolites and enzymes might leak into the bloodstream. Co-treatment with DOX–SAE significantly restored these oxidants/antioxidants’ biomarkers. These data indicate the promising role of SAE in enhancing the impact of antioxidants/hemostasis during DOX treatment in rats. The increased GSH levels resulting from SAE treatment helped to eliminate active DOX metabolites and scavenge free radicals involved in lipid oxidation, reversing hepatic damage. Similar effects of SAE treatment have been reported in experimental animals, which confirmed the hepatoprotective action of cloves [23,24,46,47].
Ferroptosis is one of the types of cell death that have been identified in recent years. It is associated with lipid peroxidation and significant iron build up and plays a role in several pathophysiological processes [48]. The results show that DOX injection increased the hepatic iron content in rats compared with that in the control group, suggesting the increased storage of hepatic iron, which in turn induced the generation of ROS and lipid peroxide, thereby leading to ferroptosis [49]. GPX-4 protects cells against lipid peroxidation, and the depletion of GPX-4 is one of the main initiators of ferroptosis [50]. For the first time, our investigation found that DOX decreased the hepatic levels of GSH and GPX-4 in rats in a NRF2-dependent manner, which could promote ferroptosis. Treatment with SAE led to a significant increase in hepatic GPX-4 expression, suggesting the effect of SAE in reversing ferroptosis induced by DOX injection (see Table 6 and Figure 8). SLC7A-11 can maintain intracellular GSH levels, and its reduction will inhibit GPX-4 expression. The SLC7A-11–GPX-4 pathway is an important regulatory pathway in ferroptosis, and the inhibition of the SLC7A-11–GPX-4 pathway could indicate the occurrence of ferroptosis [51]. In the current study, the hepatic SLC7A-11 levels were decreased after DOX poisoning; this finding supports the conclusion that the inhibition of the SLC7A-11–GPX-4 pathway may have negative effects on DOX intoxication in rats. In contrast, SAE treatment led to a significant increase in the protein level of SLC7A-11.
Interestingly, FTH-1, a key subunit of ferritin, is involved in signaling pathways in several diseases and in maintaining the cellular iron balance during ferroptosis. The process is mediated by NCOA-4, which selectively binds FTH-1, resulting in iron release [52]. High iron concentrations inhibit the binding of FTH-1 to NCOA-4 and enhance the degradation of NCOA-4, leading to the inhibition of ferritin degradation and ferroptosis [53]. In this study, the hepatic FTH-1 expression level was significantly downregulated in the DOX-injected group, while the NCOA-4 expression level was significantly improved. However, concomitant treatment with DOX–SAE significantly restored FTH-1 and NCOA-4 levels in the liver tissues of treated rats. These data suggest that SAE could be an FTH-1 inducer, a potent ferroptosis inhibitor that could be used as a pharmacological target for inhibiting ferroptosis induced by DOX. The current investigation reports for the first time a new mechanism of action of SAE that could ameliorate DOX-induced hepatotoxicity by targeting ferroptosis in rats. Additionally, DOX causes inflammation, as seen in the increased production of several proinflammatory cytokines in liver tissues [54]. Consequently, limiting inflammation might be a useful tactic to stop DOX hepatotoxicity. The present results show that the hepatic levels of inflammatory mediators, including IL-6, IL-1β, TNF-α, NF-κB, and COX-2, were significantly increased following DOX injection in rats. The increased level of these pro-inflammatory mediators in sequence leads to more hepatic injuries due to DOX exposure. However, these inflammatory cytokines were significantly restored in the DOX–SAE-treated group, and this finding suggests that the immunoregulatory properties of SAE acted against the inflammatory responses that were promoted by DOX injection in rats (see Table 7). These results may underline the mechanism of the anti-inflammatory effects exhibited by SAE. These findings are in accordance with previous studies that demonstrated the anti-inflammatory role of plant extracts against DOX-induced hepatic inflammation in experimental animals [4,55,56]. A previous study reported that Mokko lactone attenuated DOX-induced hepatotoxicity in rats through suppressing hepatic inflammatory infiltration [43]. Furthermore, Sandamali et al. (2022) reported that Nauclea orientalis (L.) bark extract protected rats from DOX-induced inflammation [57]. Cytochrome enzymes convert DOX into doxorubicinol and other hazardous byproducts that can damage the liver [43]. Histopathological changes affecting liver tissues, including hepatocyte vacuolation, have been linked to DOX treatment, according to a prior publication supporting the previous biochemical results [3]. In concordance with previous reports, our histopathological investigations of the liver section of DOX-injected rats showed extensive hepatocyte degeneration with a dilated central vein. The liver section of the DOX–SAE-treated group represented a significant improvement in the hepatic architecture (see Figure 10). These results suggest the beneficial role of SAE treatment in restoring hepatic cell damage that was promoted by DOX injection [3,4,43,44,56]. A previous study demonstrated that an S. aromaticum fraction rich in eugenol reversed biochemical and histopathological alterations in liver cirrhosis and suppressed hepatic cell injury [46].

4. Materials and Methods

4.1. Chemicals

Ethyl alcohol, ferric chloride (FeCl3), aluminum chloride (AlCl3), sodium nitroprusside, trichloroacetic acid (TCA), potassium ferricyanide (K3Fe3(CN)6), and gallic acid were purchased from Algomhoria Co., 23 El Sawah St., El Amiriya, Cairo, Egypt. Butyl hydroxy toluene (BHT) and 2,2-diphenyl-2-picrylhydrazyl (DPPH) (catalog no. 300267) were procured from Merck Co., Mumbai 400079, Maharashtra, India. Doxorubicin hydrochloride (catalog no. D5220, 98–102% HPLC) was purchased from Sigma-Aldrich (Oakville, ON L6H 6J8, Canada).

4.2. Collection and Preparation of Plant Materials

S. aromaticum flower buds were purchased from the Carrefour Market in Tanta, Egypt in November 2023. The plant was authenticated by a specialist and complied with the institutional guidelines. The chopped buds were ground into a powder, 50 g of the powdered flowers in 500 mL of ethanol (70%) were filtered, and S. aromaticum extract (SAE) was obtained [58].

4.3. Phytochemicals Analysis of SAE

The phytochemicals and DPPH radical scavenging capability were evaluated in the SAE. Butyl hydroxytoluene (BHT) was used as a standard antioxidant [59,60,61,62].

4.4. Gas Chromatography and Mass Spectrometry (GC-MS) Profiling of SAE

A Trace GC 1310-ISQ mass spectrometer (Thermo Scientific, Austin, TX, USA) was used to identify the phytochemicals in the SAE. The components were detected, and their mass spectral and retention periods were matched to those found in the mass spectral databases of WILEY 09 and NIST 11 [63].

4.5. Molecular Docking Analysis

The SMILES codes of the compounds doxorubicin and eugenol were used as the input for the ADMETlab 2.0 web server to predict their pharmacokinetic properties, including the absorption, distribution, metabolism, excretion, and toxicity (ADMET) [64]. The protein structures for NRF-2 (Uniprot ID: O54968), SLC7A-11 (Uniprot ID: D4ADU2), and GPX-4 (Uniprot ID: P36970) were collected from the UniProt database, and models were generated using the AlphaFold server [65]. The active sites of all proteins were predicted using the CB-Dock2 server, which employs deep learning algorithms to identify potential binding pockets and catalytic sites. The prepared protein structures were further processed using AutoDock Tools 1.5.7. The active constituents obtained from the ligand molecules were retrieved from the PubChem database in their respective SDF formats. These ligands were then minimized using the Avogadro 1.2.0 software, employing the Force Field algorithm (MMFF94) and the Conjugate Gradients algorithm [66,67]. The minimized ligand structures were converted to the PDBQT format compatible with AutoDock Vina. The search space for the docking simulations was defined based on the predicted active site regions obtained from the deep site server. The docking results were visualized and analyzed using BIOVIA Discovery Studio 2020 (San Diego, CA, USA).

4.6. Oral Median Lethal Dose (LD50) of SAE

The oral LD50 following SAE administration was evaluated in rats. Rats were gavaged with SAE (1–5 g/kg b. wt.) then observed to see if there were any signs of toxicity for 24 h. The LD50 value was determined by the probit analysis [68].

4.7. Animals and Experimental Design

Forty adult male Sprague Dawley rats (130–150 g, 5–6 weeks of age) were purchased from Helwan University, Egypt. Male rats were chosen in order to avoid the fluctuating hormone levels and the menstrual cycle that could make the data difficult to interpret and the results more variable. Our study was conducted in accordance with the Faculty of Science, Tanta University animal care committee (IACUC-SCI-TU-0321). Rats were divided into four groups (n = 10). G1 was a negative control group that was injected i.p. with saline daily, G2 was orally given 1/10 of SAE LD50 (340 mg/kg) daily for a month, G3 was injected with 4 mg/kg of DOX i.p. once a week for a month [69], and G4 received the same DOX injection as G3 and the same SAE administration as G2. Serum and liver tissues were collected for biochemical, molecular, and histopathological investigations.

4.8. Biochemical Analysis

Alanine aminotransferase (ALT) (catalog no. AL103145), aspartate aminotransferase (AST) (catalog no. AS106145), alkaline phosphatase (ALP) (catalog no. AP1020), total bilirubin (catalog no. BR1111), and direct bilirubin (catalog no. BR1112) were assessed using colorimetric kits (Spectrum Diagnostics, Egypt). Hepatic levels of malondialdehyde (MDA) (catalog no. MD2529), superoxide dismutase (SOD) (catalog no. SD2521), catalase (CAT) (catalog no. CA2517), and reduced glutathione (GSH) (catalog no. GR2511) were measured by using their respective kits (Biodiagnostic, Giza Governorate, Egypt). The protein concentration was measured by the method of Lowry et al. (1951) using bovine serum albumin (BSA) as a standard [70]. Iron levels in rats’ livers were detected using the iron assay kit (catalog no. ab83366). Hepatic nuclear factor erythroid 2-related factor 2 (NRF-2) (catalog no. MBS752046), solute carrier family 7, member 11 (SLC7A-11) (catalog no. MBS2705481), glutathione peroxidase (GPX-4) (catalog no. MBS934198), ferritin heavy chain-1 (FTH-1) (catalog no. MBS2886777), acyl-CoA synthetase long-chain family member 4 (ACSL-4) (catalog no. MBS9903690), and nuclear receptor coactivator 4 (NCOA-4) (catalog no. MBS7269622) were determined using the respective rat-specific ELISA kits from MyBioSource, Inc., San Diego, CA, USA. Rat-specific ELISA kits were used for the measurement of the inflammatory biomarkers, including interleukin-6 (IL-6) (catalog no. E-HSEL-R0004), interleukin-1β (IL-1β) (catalog no. E-EL-R0012), tumor necrosis factor-α (TNF-α) (catalog no. RAB0479), nuclear factor kappa-B (NF-κB) (catalog no. MBS453975), and cyclooxygenase-2 (COX-2) (catalog no. MBS266603) in the liver homogenates of the different groups.

4.9. Molecular Analysis

The mRNA expression of the NRF2, SLC7A11, GPX4, FTH1, and NCOA4 genes was evaluated in liver tissues using GAPDH as an internal reference. The primers were prepared using the Primer-Blast program from NCBI (Table 9). The relative expression of target genes was estimated [71].

4.10. Histopathological Investigations

Liver tissues were sectioned at 5 μm, embedded in paraffin wax, washed in xylene, and then sliced and fixed in 10% buffered formalin. Hematoxylin and eosin (H&E) staining was applied to the sections, which were then viewed under an Olympus CX31 light microscope and captured on a digital camera (Olympus Camedia 5060, Tokyo, Japan) [72]. Hepatic damage was analyzed based on the severity and tissue damage percentage. A scale (0–4) was used, where 0 is normal tissue, 1 is <25% hepatic tissue damage, 2 is 26–50% hepatic tissue damage, 3 is 51–75% hepatic tissue damage, and 4 is >75% hepatic tissue damage [73].

4.11. Statistical Analysis

A one-way analysis of variance (ANOVA) was used to assess significant variations. The software GraphPad Prism, Inside Scientific Co., (San Diego, CA, USA), https://www.graphpad.com/ (accessed on 5 April 2024) was utilized for the evaluation of results. For multiple comparisons, Tukey’s test was applied, and statistical significance was established at p < 0.05.

5. Conclusions

SAE may be a highly effective protective agent against hepatotoxicity brought on by DOX therapy. It has this effect through its inhibition of oxidative stress and inflammation and its upregulation of the NRF-2–SLC7A-11–GPX-4 signaling pathway. Further preclinical and clinical studies should investigate the effects of SAE against other toxicities induced by DOX.

Author Contributions

Conceptualization, supervision, funding acquisition, design, writing—original draft preparation, and editing, A.M.A., A.S.A.-A., M.A.A. and K.S.E.-S.; investigation, methodology, resources, and formal analysis, A.A.A., N.Y.A., S.A. and S.A.A.; validation, investigation, methodology, and writing—original draft preparation, M.A.M., T.A., I.A. and T.M.E. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by a grant from the Princess Nourah bint Abdulrahman University Researchers Supporting Project number (PNURSP2024R227), Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki, and the study was approved by the animal care and use committee, Faculty of Science, Tanta University (IACUC-SCI-TU-0321), Egypt.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author. The data are not publicly available due to ethical restrictions, our research still be used in ongoing studies, future research, and sharing it prematurely could jeopardize the intellectual property or future publications.

Acknowledgments

The authors extend their appreciation to the Princess Nourah bint Abdulrahman University Researchers Supporting Project number (PNURSP2024R227), Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia for funding this work.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Li, S.; Yuan, S.; Zhao, Q.; Wang, B.; Wang, X.; Li, K. Quercetin enhances chemotherapeutic effect of doxorubicin against human breast cancer cells while reducing toxic side effects of it. Biomed. Pharmacother. 2018, 100, 441–447. [Google Scholar] [CrossRef]
  2. Damodar, G.; Smitha, T.; Gopinath, S.; Vijayakumar, S.; Rao, Y. An evaluation of hepatotoxicity in breast cancer patients receiving injection doxorubicin. Ann. Med. Health Sci. Res. 2014, 4, 74–79. [Google Scholar] [CrossRef]
  3. Reddy, A.; Anjaneyulu, Y.; Shivakumar, P.; Rani, M. A study on the toxic effects of doxorubicin on the histology of certain organs. Toxicol. Int. 2012, 19, 241–244. [Google Scholar] [CrossRef]
  4. El-Said, K.S.; Haidyrah, A.S.; Mobasher, M.A.; Khayyat, A.I.A.; Shakoori, A.; Al-Sowayan, N.S.; Barnawi, I.O.; Mariah, R.A. Artemisia annua extract attenuate doxorubicin-induced hepatic injury via PI-3K/Akt/Nrf-2-mediated signaling pathway in rats. Int. J. Mol. Sci. 2023, 24, 15525. [Google Scholar] [CrossRef] [PubMed]
  5. Wu, Z.; Zhu, Y.; Liu, W.; Balasubramanian, B.; Xu, X.; Yao, J.; Lei, X. Ferroptosis in liver disease: Natural active compounds and therapeutic implications. Antioxidants 2024, 13, 352. [Google Scholar] [CrossRef]
  6. Chen, J.; Li, X.; Ge, C.; Min, J.; Wang, F. The multifaceted role of ferroptosis in liver disease. Cell Death Differ. 2022, 29, 467–480. [Google Scholar] [CrossRef] [PubMed]
  7. Wang, X.; Zhou, Y.; Min, J.; Wang, F. Zooming in and out of ferroptosis in human disease. Front. Med. 2023, 17, 173–206. [Google Scholar] [CrossRef] [PubMed]
  8. Dong, H.; Xia, Y.; Jin, S.; Xue, C.; Wang, Y.; Hu, R.; Jiang, H. Nrf2 attenuates ferroptosis-mediated IIR-ALI by modulating TERT and SLC7A11. Cell Death Dis. 2021, 12, 1027. [Google Scholar] [CrossRef] [PubMed]
  9. Ji, X.; Qian, J.; Rahman, S.M.J.; Siska, P.J.; Zou, Y.; Harris, B.K.; Hoeksema, M.D.; Trenary, I.A.; Heidi, C.; Eisenberg, R.; et al. xCT (SLC7A11)-mediated metabolic reprogramming promotes non-small cell lung cancer progression. Oncogene 2018, 37, 5007–5019. [Google Scholar] [CrossRef]
  10. Anandhan, A.; Dodson, M.; Schmidlin, C.J.; Liu, P.; Zhang, D.D. Breakdown of an ironclad defense system: The critical role of nrf2 in mediating ferroptosis. Cell Chem. Biol. 2020, 27, 436–447. [Google Scholar] [CrossRef]
  11. Wu, L.; Zhang, Y.; Wang, G.; Ren, J. Molecular mechanisms and therapeutic targeting of ferroptosis in doxorubicin-induced car-diotoxicity. JACC Basic Transl. Sci. 2024, 9, 811–826. [Google Scholar] [CrossRef] [PubMed]
  12. Zhao, T.; Yu, Z.; Zhou, L.; Wang, X.; Hui, Y.; Mao, L.; Fan, X.; Wang, B.; Zhao, X.; Sun, C. Regulating Nrf2-GPx4 axis by bicyclol can prevent ferroptosis in carbon tetrachloride-induced acute liver injury in mice. Cell Death Discov. 2022, 8, 380. [Google Scholar] [CrossRef] [PubMed]
  13. Dixon, S.J.; Lemberg, K.M.; Lamprecht, M.R.; Skouta, R.; Zaitsev, E.M.; Gleason, C.E.; Patel, D.N.; Bauer, A.J.; Cantley, A.M.; Yang, W.S.; et al. Ferroptosis: An iron-dependent form of non-apoptotic cell death. Cell 2012, 149, 1060–1072. [Google Scholar] [CrossRef] [PubMed]
  14. De la Torre, B.G.; Albericio, F. The pharmaceutical industry in 2018. An analysis of FDA drug approvals from the perspective of molecules. Molecules 2019, 24, 809. [Google Scholar] [CrossRef] [PubMed]
  15. Patel, N.; Joseph, C.; Corcoran, G.B.; Ray, S.D. Silymarin modulates doxorubicin-induced oxidative stress, Bcl-xL and p53 expression while preventing apoptotic and necrotic cell death in the liver. Toxicol. Appl. Pharmacol. 2010, 245, 143–152. [Google Scholar] [CrossRef]
  16. Mahmoudi, F.; Arasteh, O.; Elyasi, S. Preventive and therapeutic use of herbal compounds against doxorubicin induced hepato-toxicity: A comprehensive review. Naunyn Schmiedebergs Arch. Pharmacol. 2023, 396, 1595–1617. [Google Scholar] [CrossRef] [PubMed]
  17. Xi, Y.; Chi, Z.; Tao, X.; Zhai, X.; Zhao, Z.; Ren, J.; Yang, S.; Dong, D. Naringin against doxorubicin-induced hepatotoxicity in mice through reducing oxidative stress, inflammation, and apoptosis via the up-regulation of SIRT1. Environ. Toxicol. 2023, 38, 1153–1161. [Google Scholar] [CrossRef]
  18. Nugraha, S.E.; Yuandani, Y.; Syahputra, R.A. Protective activity of beetroot extract on doxorubicin-induced hepatic and renal tox-icity in rat model. Maced. J. Med. Sci. 2021, 9, 1037–1042. [Google Scholar] [CrossRef]
  19. Mete, R.; Oran, M.; Topcu, B.; Oznur, M.; Seber, E.S.; Gedikbasi, A.; Yetisyigit, T. Protective effects of onion (Allium cepa) extract against doxorubicin-induced hepatotoxicity in rats. Toxicol. Ind. Health 2013, 32, 551–557. [Google Scholar] [CrossRef]
  20. Kaur, K.; Kaushal, S. Phytochemistry and pharmacological aspects of Syzygium aromaticum: A review. J. Pharmacogn. Phyto-Chem. 2019, 8, 398–404. [Google Scholar]
  21. Pandey, V.K.; Srivastava, S.; Ashish Dash, K.K.; Singh, R.; Dar, A.H.; Singh, T.; Farooqui, A.; Shaikh, A.M.; Kovacs, B. Bioactive properties of clove (Syzygium aromaticum) essential oil nanoemulsion: A comprehensive review. Heliyon 2024, 10, e22437. [Google Scholar] [CrossRef] [PubMed]
  22. Mergulhão, N.L.O.N.; Bulhões, L.C.G.; Silva, V.C.; Duarte, I.F.B.; Basílio-Júnior, I.D.; Freitas, J.D.; Oliveira, A.J.; Goulart, M.O.F.; Barbosa, C.V.; Araújo-Júnior, J.X. Insights from Syzygium aromaticum essential oil: Encapsulation, characterization, and antioxidant activity. Pharmaceuticals 2024, 17, 599. [Google Scholar] [CrossRef] [PubMed]
  23. Pourlak, T.; Halimi, M.; Pourlak, T.; Maroufi, P.; Ghaderpour, S.; Shokoohi, A. Effect of Extracts of Cloves (Syzygium Aromaticum) on Hepatic Cell Damage and Oxidative Stress Caused by Diabetes in Adult Rats. Intern. Med. Today 2020, 26, 432–447. [Google Scholar] [CrossRef]
  24. Abdelrahman, M.T.; Maina, E.N.; Elshemy, H.A. Clove (Syzygium aromaticum) and honey extracts significantly reduce inflammatory cytokines and liver function enzymes in experimental rats fed on carbon tetrachloride (CCl4). J. Radiat. Res. Appl. Sci. 2018, 11, 416–422. [Google Scholar] [CrossRef]
  25. Adegbola, M.V.; Anyim, G.; Ntwasa, M.; Ayeleso, A.O.; Oyedepo, T.A. Potential Effect of Syzygium aromaticum (Cloves) Extract on Serum Antioxidant Status and Lipid Profiles in Wistar Rats with Artesunate Toxicity. Appl. Sci. 2022, 12, 8216. [Google Scholar] [CrossRef]
  26. Agboola, J.B.; Ehigie, A.F.; Ehigie, L.O.; Ojeniyi, F.D.; Olayemi, A.A. Ameliorative role of Syzygium aromaticum aqueous extract on synaptosomal tyrosine hydroxylase activity, oxidative stress parameters, and behavioral changes in lead-induced neurotoxicity in mice. J. Food Biochem. 2022, 46, e14115. [Google Scholar] [CrossRef]
  27. Wu, Q.; Chen, Z.; Ding, Y.; Tang, Y.; Cheng, Y. Protective effect of traditional Chinese medicine on non-alcoholic fatty liver disease and liver cancer by targeting ferroptosis. Front. Nutr. 2022, 9, 1033129. [Google Scholar] [CrossRef]
  28. Batiha, G.E.; Alkazmi, L.M.; Wasef, L.G.; Beshbishy, A.M.; Nadwa, E.H.; Rashwan, E.K. Syzygium aromaticum L. (Myrtaceae): Tradi-tional uses, bioactive chemical constituents, pharmacological and toxicological activities. Biomolecules 2020, 10, 202. [Google Scholar] [CrossRef]
  29. Liu, J.; Huang, C.; Liu, J.; Meng, C.; Gu, Q.; Du, X.; Yan, M.; Yu, Y.; Liu, F.; Xia, C. Nrf2 and its dependent autophagy activation cooperatively counteract ferroptosis to alleviate acute liver injury. Pharmacol. Res. 2023, 187, 106563. [Google Scholar] [CrossRef]
  30. Li, X.; Ma, N.; Xu, J.; Zhang, Y.; Yang, P.; Su, X.; Xing, Y.; An, N.; Yang, F.; Zhang, G.; et al. Targeting Ferroptosis: Pathological Mechanism and Treatment of Ischemia-Reperfusion Injury. Oxidative Med. Cell. Longev. 2021, 2021, 1587922. [Google Scholar] [CrossRef]
  31. Qiu, S.; Li, X.; Zhang, J.; Shi, P.; Cao, Y.; Zhuang, Y.; Tong, L. Neutrophil membrane-coated taurine nanoparticles protect against hepatic ischemia-reperfusion injury. Eur. J. Pharmacol. 2023, 949, 175712. [Google Scholar] [CrossRef] [PubMed]
  32. Lone, Z.A.; Jain, N.K. Phytochemical analysis of Clove (Syzygium aromaticum) dried flower buds extract and its therapeutic importance. J. Drug Deliv. Ther. 2022, 12, 87–92. [Google Scholar] [CrossRef]
  33. Abdul Aziz, A.H.; Rizkiyah, D.N.; Qomariyah, L.; Irianto, I.; Che Yunus, M.A.; Putra, N.R. Unlocking the full potential of Clove (Syzygium aromaticum) spice: An overview of extraction techniques, bioactivity, and future opportunities in the food and beverage industry. Processes 2023, 11, 2453. [Google Scholar] [CrossRef]
  34. Zhou, S.; Liu, Y.; Wang, S.; Wang, L. Effective prediction of short hydrogen bonds in proteins via machine learning method. Sci. Rep. 2022, 12, 469. [Google Scholar] [CrossRef] [PubMed]
  35. Atkovska, K.; Samsonov, S.A.; Paszkowski-Rogacz, M.; Pisabarro, M.T. Multipose binding in molecular docking. Int. J. Mol. Sci. 2014, 15, 2622–2645. [Google Scholar] [CrossRef] [PubMed]
  36. Mili, A.; Das, S.; Nandakumar, K.; Lobo, R. Molecular docking and dynamics guided approach to identify potential anti-inflammatory molecules as NRF2 activator to protect against drug-induced liver injury (DILI): A computational study. J. Biomol. Struct. Dyn. 2023, 41, 9193–9210. [Google Scholar] [CrossRef] [PubMed]
  37. Chen, D.; Oezguen, N.; Urvil, P.; Ferguson, C.; Dann, S.M.; Savidge, T.C. Regulation of protein-ligand binding affinity by hydrogen bond pairing. Sci. Adv. 2016, 2, e1501240. [Google Scholar] [CrossRef]
  38. Eliaa, S.G.; Al-Karmalawy, A.A.; Saleh, R.M.; Elshal, M.F. Empagliflozin and doxorubicin synergistically inhibit the survival of triple-negative breast cancer cells via interfering with the mTOR pathway and inhibition of calmodulin: In vitro and molecular docking studies. ACS Pharmacol. Transl. Sci. 2020, 3, 1330–1338. [Google Scholar] [CrossRef]
  39. Prasanna, P.L.; Renu, K.; Valsala Gopalakrishnan, A. New molecular and biochemical insights of doxorubicin-induced hepatotoxicity. Life Sci. 2020, 250, 117599. [Google Scholar] [CrossRef]
  40. Nirmala, J.; John, S.; Sumanth Raj, U.; Narasa, S.; Nagarajan, R. Chapter 39—Toxicity of clove (Syzygium aromaticum) extract. Acad. Press. 2022, 39, 663–674. [Google Scholar]
  41. Liu, B.B.; Luo, L.; Liu, X.L.; Geng, D.; Li, C.F.; Chen, S.M.; Chen, X.M.; Yi, L.T.; Liu, Q. Essential oil of Syzygium aromaticum reverses the deficits of stress-induced behaviors and hippocampal p-ERK/p-CREB/brain-derived neurotrophic factor expression. Planta Medica 2015, 81, 185–192. [Google Scholar] [CrossRef] [PubMed]
  42. Liao, W.; Rao, Z.; Wu, L.; Chen, Y.; Li, C. Cariporide Attenuates Doxorubicin-Induced Cardiotoxicity in Rats by Inhibiting Oxidative Stress, Inflammation and Apoptosis Partly Through Regulation of Akt/GSK-3β and Sirt1 Signaling Pathway. Front. Pharmacol. 2022, 13, 850053. [Google Scholar] [CrossRef] [PubMed]
  43. Sirwi, A.; Shaik, R.A.; Alamoudi, A.J.; Eid, B.G.; Kammoun, A.K.; Ibrahim, S.R.M.; Mohamed, G.A.; Abdallah, H.M.; Abdel-Naim, A.B. Mokko lactone attenuates doxorubicin-induced hepatotoxicity in rats: Emphasis on Sirt-1/FOXO1/NF-κB axis. Nutrients 2021, 13, 4142. [Google Scholar] [CrossRef] [PubMed]
  44. Ahmed, O. Camellia sinensis and epicatechin abate doxorubicin-induced hepatotoxicity in male Wistar rats via their modulatory effects on oxidative stress, inflammation, and apoptosis. J. App. Pharmaceut. Sci. 2019, 9, 30–44. [Google Scholar]
  45. AlAsmari, A.F.; Alharbi, M.; Alqahtani, F.; Alasmari, F.; AlSwayyed, M.; Alzarea, S.I.; Al-Alallah, I.A.; Alghamdi, A.; Hakami, H.M.; Alyousef, M.K.; et al. Diosmin Alleviates Doxorubicin-Induced Liver Injury via Modulation of Oxidative Stress-Mediated Hepatic Inflammation and Apoptosis via NfkB and MAPK Pathway: A Preclinical Study. Antioxidants 2021, 10, 1998. [Google Scholar] [CrossRef]
  46. Ali, S.; Prasad, R.; Mahmood, A.; Routray, I.; Shinkafi, T.S.; Sahin, K.; Kucuk, O. Eugenol-rich fraction of Syzygium aromaticum (Clove) reverses biochemical and histopathological changes in liver cirrhosis and inhibits hepatic cell proliferation. J. Cancer Prev. 2014, 19, 288–300. [Google Scholar] [CrossRef]
  47. Abd Al-azem, D.; Al-Derawi, K.H.; Al-Saadi, S. The protective effects of Syzygium aromaticum essential oil extract against metho-trexate induced hepatic and renal toxicity in rats. Pure App. Microbiol. 2019, 13, 505–515. [Google Scholar] [CrossRef]
  48. Li, J.; Cao, F.; Yin, H.L.; Huang, Z.J.; Lin, Z.T.; Mao, N.; Sun, B.; Wang, G. Ferroptosis: Past, present and future. Cell Death Dis. 2020, 11, 88. [Google Scholar] [CrossRef]
  49. Lin, R.; Jia, Z.; Chen, H.; Xiong, H.; Bian, C.; He, X.; Wei, B.; Fu, J.; Zhao, M.; Li, J. Ferrostatin-1 alleviates liver injury via decreasing ferroptosis following ricin toxin poisoning in rat. Toxicology 2024, 503, 153767. [Google Scholar] [CrossRef]
  50. Xu, S.; Li, X.; Li, Y.; Li, X.; Lv, E.; Zhang, X.; Shi, Y.; Wang, Y. Neuroprotective effect of Dl-3-n-butylphthalide against ische-mia-reperfusion injury is mediated by ferroptosis regulation via the SLC7A11/GSH/GPX4 pathway and the attenuation of blood-brain barrier disruption. Front. Aging Neurosci. 2023, 15, 1028178. [Google Scholar] [CrossRef]
  51. Wu, Y.; Jia, Q.; Tang, Q.; Deng, H.; He, Y.; Tang, F. Berberine-mediated Ferroptosis through System Xc-/GSH/GPX4 Axis Inhibits Metastasis of Nasopharyngeal Carcinoma. J. Cancer 2024, 15, 685–698. [Google Scholar] [CrossRef] [PubMed]
  52. Santana-Codina, N.; Mancias, J.D. The Role of NCOA4-Mediated Ferritinophagy in Health and Disease. Pharmaceuticals 2018, 11, 114. [Google Scholar] [CrossRef] [PubMed]
  53. Latunde-Dada, G.O. Ferroptosis: Role of lipid peroxidation, iron and ferritinophagy. Biochim. Biophys. Acta (BBA) Gen. Subj. 2017, 1861, 1893–1900. [Google Scholar] [CrossRef] [PubMed]
  54. Song, S.; Chu, L.; Liang, H.; Chen, J.; Liang, J.; Huang, Z.; Zhang, B.; Chen, X. Protective effects of dioscin against doxorubicin-induced hepatotoxicity via regulation of Sirt1/FOXO1/NF-κb signal. Front. Pharmacol. 2019, 10, 1030. [Google Scholar] [CrossRef] [PubMed]
  55. Alherz, F.A.; Negm, W.A.; El-Masry, T.A.; Elmorshedy, K.E.; El-Kadem, A.H. The potential beneficial role of Ginkgetin in doxo-rubicin-induced hepatotoxicity: Elucidating the underlying claim. Biomed. Pharmacother. 2023, 165, 115010. [Google Scholar] [CrossRef]
  56. Li, Q.; Li, X.; Zhou, Z.; Zhu, P.; Tuo, N.; Ge, J.; Liu, Z.; Chen, D. Kaempferide Inhibits DOX-induced Liver Inflammation by Activating AMPKα/SIRT1. Pharmacogn. Mag. 2024, 20, 1304–1312. [Google Scholar] [CrossRef]
  57. Sandamali, J.A.N.; Hewawasam, R.P.; Jayatilaka, K.A.P.W.; Mudduwa, L.K.B. Nauclea orientalis (L.) bark extract protects rat car-diomyocytes from doxorubicin-induced oxidative stress, inflammation, apoptosis, and DNA fragmentation. Oxid. Med. Cell Longev. 2022, 2022, 1714841. [Google Scholar] [CrossRef]
  58. Mobasher, M.A.; Alsirhani, A.M.; Alwaili, M.A.; Baakdah, F.; Eid, T.M.; Alshanbari, F.A.; Alzahri, R.Y.; Alkhodair, S.A.; El-Said, K.S. Annona squamosa fruit extract ameliorates lead acetate-induced testicular injury by modulating JAK-1/STAT-3/SOCS-1 signaling in male rats. Int. J. Mol. Sci. 2024, 25, 5562. [Google Scholar] [CrossRef]
  59. Singleton, V.L.; Orthofer, R.; Lamuela-Raventós, R.M. Analysis of total phenols and other oxidation substrates and antioxidants by means of folin-ciocalteu reagent. Methods Enzymol. 1999, 299, 152–178. [Google Scholar] [CrossRef]
  60. Prieto, P.; Pineda, M.; Aguilar, M. Spectrophotometric quantitation of antioxidant capacity through the formation of a phospho-molybdenum complex: Specific application to the determination of vitamin E. Anal. Biochem. 1999, 269, 337–341. [Google Scholar] [CrossRef]
  61. Hiai, S.; Oura, H.; Odaka, Y.; Nakajima, T. A colorimetric estimation of ginseng saponins. Planta Medica 1975, 28, 363–369. [Google Scholar] [CrossRef] [PubMed]
  62. Blois, M.S. Antioxidant Determinations by the Use of a Stable Free Radical. Nature 1958, 181, 1199–1200. [Google Scholar] [CrossRef]
  63. El-Said, K.S.; Hussein, S.; Alrashdi, B.M.; Mahmoud, H.A.; Ibrahim, M.A.; Elbakry, M.; El-Tantawy, H.; Kabil, D.I.; El-Naggar, S.A. Musa sp. Leaves Extract Ameliorates the Hepato-Renal Toxicities Induced by Cadmium in Mice. Molecules 2022, 27, 559. [Google Scholar] [CrossRef] [PubMed]
  64. Xiong, G.; Wu, Z.; Yi, J.; Fu, L.; Yang, Z.; Hsieh, C.; Yin, M.; Zeng, X.; Wu, C.; Lu, A.; et al. ADMETlab 2.0: An integrated online platform for accurate and comprehensive predictions of ADMET properties. Nucleic Acids Res. 2021, 49, W5–W14. [Google Scholar] [CrossRef] [PubMed]
  65. Cheng, J.; Novati, G.; Pan, J.; Bycroft, C.; Žemgulytė, A.; Applebaum, T.; Pritzel, A.; Wong, L.H.; Zielinski, M.; Sargeant, T.; et al. Accurate proteome-wide missense variant effect prediction with AlphaMissense. Science 2023, 381, 1303. [Google Scholar] [CrossRef]
  66. Hanwell, M.D.; Curtis, D.E.; Lonie, D.C.; Vandermeersch, T.; Zurek, E.; Hutchison, G.R. Avogadro: An advanced semantic chemical editor, visualization, and analysis platform. J. Cheminform. 2012, 4, 17. [Google Scholar] [CrossRef]
  67. Trott, O.; Olson, A.J. AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J. Comput. Chem. 2010, 31, 455–461. [Google Scholar] [CrossRef]
  68. Finney, D.J.; Stevens, W.L. A table for the calculation of working probits and weight in propit analysis. Biometrika 1948, 35, 191–201. [Google Scholar] [CrossRef]
  69. Warpe, V.S.; Mali, V.R.; Arulmozhi, S.; Bodhankar, S.L.; Mahadik, K.R. Cardioprotective effect of ellagic acid on doxorubicin induced cardiotoxicity in wistar rats. J. Acute Med. 2015, 5, 1–8. [Google Scholar] [CrossRef]
  70. Lowry, O.H.; Rosebrough, N.J.; Farr, A.L.; Randall, R.J. Protein measurement with the Folin Phenol reagent. J. Biol. Chem. 1951, 193, 265–275. [Google Scholar] [CrossRef]
  71. Livak, K.J.; Schmittgen, T.D. Analysis of relative gene expression data using real-time quantitative PCR and the 2−∆∆CT method. Methods 2001, 25, 402–408. [Google Scholar] [CrossRef] [PubMed]
  72. Bancroft, J.; Gamble, M. Theory and Practice of Histological Techniques; Elsevier Health Sciences: Philadelphia, PA, USA, 2008. [Google Scholar]
  73. Orabi, S.H.; Abd Eldaium, D.; Hassan, A.; Sabagh, H.S.E.; Abd Eldaim, M.A. Allicin modulates diclofenac sodium induced hepa-tonephro toxicity in rats via reducing oxidative stress and caspase 3 protein expression. Environ. Toxicol. Pharmacol. 2020, 74, 103306. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The abundant bioactive compounds in SAE.
Figure 1. The abundant bioactive compounds in SAE.
Ijms 25 12541 g001
Figure 2. Radar chart for ADMET screening showing the upper, lower, and predicted values of various physicochemical and molecular properties of DOX (A), eugenol (B), and 10-Heptadecen-8-ynoic acid, methyl ester (C). MW, molecular weight; nRig, number of rigid bonds; fChar, formal charge; nHet, number of heteroatoms; MaxRing, number of atoms in the biggest ring; nRing, number of rings; nRot, number of rotatable bonds; TPSA, topological polar surface area; nHD, number of hydrogen bond donors; nHA, number of hydrogen bond acceptors; LogP, Log of the octanol–water partition coefficient; LogS, Log of the aqueous solubility; LogD, Log at physiological pH (7.4).
Figure 2. Radar chart for ADMET screening showing the upper, lower, and predicted values of various physicochemical and molecular properties of DOX (A), eugenol (B), and 10-Heptadecen-8-ynoic acid, methyl ester (C). MW, molecular weight; nRig, number of rigid bonds; fChar, formal charge; nHet, number of heteroatoms; MaxRing, number of atoms in the biggest ring; nRing, number of rings; nRot, number of rotatable bonds; TPSA, topological polar surface area; nHD, number of hydrogen bond donors; nHA, number of hydrogen bond acceptors; LogP, Log of the octanol–water partition coefficient; LogS, Log of the aqueous solubility; LogD, Log at physiological pH (7.4).
Ijms 25 12541 g002
Figure 3. DOX interactions with three target proteins (NRF-2, SLC7A-11, and GPX-4) (2D and 3D). (A) DOX interaction with NRF-2, the conventional H-bond (GLY280, TYR284, GLN584, and GLN584), and the hydrophobic interactions (VAL563, LEU582, and VAL590); (B) DOX interaction with SLC7A-11, the conventional H-bond (TYR251), and the hydrophobic interactions (ILE52 and LYS43); (C) DOX interaction with GPX-4, the conventional H-bond (MET28, SER40, ARG60, GLU43, GLY26, CYS37, and LYS595), the carbon H-bond (ARG39 and GLU43), the Pi–Anion interaction (GLU43), and the Pi–Alkyl interaction (ARG39).
Figure 3. DOX interactions with three target proteins (NRF-2, SLC7A-11, and GPX-4) (2D and 3D). (A) DOX interaction with NRF-2, the conventional H-bond (GLY280, TYR284, GLN584, and GLN584), and the hydrophobic interactions (VAL563, LEU582, and VAL590); (B) DOX interaction with SLC7A-11, the conventional H-bond (TYR251), and the hydrophobic interactions (ILE52 and LYS43); (C) DOX interaction with GPX-4, the conventional H-bond (MET28, SER40, ARG60, GLU43, GLY26, CYS37, and LYS595), the carbon H-bond (ARG39 and GLU43), the Pi–Anion interaction (GLU43), and the Pi–Alkyl interaction (ARG39).
Ijms 25 12541 g003
Figure 4. Eugenol interactions with three target proteins (NRF-2, SLC7A-11, and GPX-4) (2D and 3D). (A) Eugenol interaction with NRF-2 and the hydrophobic interactions (LEU556, LEU559, LEU582, and VAL590); (B) Eugenol interaction with SLC7A-11, the conventional H-bond (ARG135 and TYR251), and the hydrophobic interactions (ILE142, ALA138, and TYR244); (C) Eugenol interaction with GPX-4, the conventional H-bond (ARG32), and the hydrophobic interactions (HIS140, ALA46, ARG32, PRO141, LYS47, ILE132, and HIS140).
Figure 4. Eugenol interactions with three target proteins (NRF-2, SLC7A-11, and GPX-4) (2D and 3D). (A) Eugenol interaction with NRF-2 and the hydrophobic interactions (LEU556, LEU559, LEU582, and VAL590); (B) Eugenol interaction with SLC7A-11, the conventional H-bond (ARG135 and TYR251), and the hydrophobic interactions (ILE142, ALA138, and TYR244); (C) Eugenol interaction with GPX-4, the conventional H-bond (ARG32), and the hydrophobic interactions (HIS140, ALA46, ARG32, PRO141, LYS47, ILE132, and HIS140).
Ijms 25 12541 g004
Figure 5. 10-Heptadecen-8-ynoic acid, methyl ester interactions with three target proteins (NRF-2, SLC7A-11, and GPX-4) (2D and 3D). (A) The interaction with NRF-2, the conventional H-bond (LYS553), and the hydrophobic interactions (PHE279, PHE287, and HIS550); (B) The interaction with SLC7A-11, the conventional H-bond (GLN240), the carbon H-bond (ARG119 and ILE120), and the hydrophobic interactions (LYS206, ALA332, LEU177, and TYR205); (C) The interaction with GPX-4, the carbon H-bond (ILE13), and the hydrophobic interactions (VAL112, ILE115, ILE142, ILE164, VAL167, ILE168, PHE161, and PHE173).
Figure 5. 10-Heptadecen-8-ynoic acid, methyl ester interactions with three target proteins (NRF-2, SLC7A-11, and GPX-4) (2D and 3D). (A) The interaction with NRF-2, the conventional H-bond (LYS553), and the hydrophobic interactions (PHE279, PHE287, and HIS550); (B) The interaction with SLC7A-11, the conventional H-bond (GLN240), the carbon H-bond (ARG119 and ILE120), and the hydrophobic interactions (LYS206, ALA332, LEU177, and TYR205); (C) The interaction with GPX-4, the carbon H-bond (ILE13), and the hydrophobic interactions (VAL112, ILE115, ILE142, ILE164, VAL167, ILE168, PHE161, and PHE173).
Ijms 25 12541 g005
Figure 6. (A) The relative body weight percentages. (B) The relative liver weight in the different groups under study. I.B.W, initial body weight; F.B.W, final body weight; SAE, Syzygium aromaticum extract; DOX, Doxorubicin. Data are expressed as mean ± S.D., n = 10. Means that do not share a letter showed a significant difference (p < 0.05).
Figure 6. (A) The relative body weight percentages. (B) The relative liver weight in the different groups under study. I.B.W, initial body weight; F.B.W, final body weight; SAE, Syzygium aromaticum extract; DOX, Doxorubicin. Data are expressed as mean ± S.D., n = 10. Means that do not share a letter showed a significant difference (p < 0.05).
Ijms 25 12541 g006
Figure 7. Hepatic malondialdehyde (MDA) (A), superoxide dismutase (SOD) (B), catalase (CAT) (C), and reduced glutathione (GSH) (D) levels in the different groups. SAE, Syzygium aromaticum extract; DOX, Doxorubicin. The values represent means ± S.D., n = 10. Means that do not share a letter showed a significant difference (p < 0.05).
Figure 7. Hepatic malondialdehyde (MDA) (A), superoxide dismutase (SOD) (B), catalase (CAT) (C), and reduced glutathione (GSH) (D) levels in the different groups. SAE, Syzygium aromaticum extract; DOX, Doxorubicin. The values represent means ± S.D., n = 10. Means that do not share a letter showed a significant difference (p < 0.05).
Ijms 25 12541 g007
Figure 8. Hepatic iron (A), nuclear factor erythroid 2-related factor 2 (NRF-2) (B), solute carrier family 7, member 11 (SLC7A-11) (C), and glutathione peroxidase (GPX-4) (D) in the different groups. SAE, Syzygium aromaticum extract; DOX, Doxorubicin. The values represent means ± S.D., n = 10. Means that do not share a letter showed a significant difference (p < 0.01).
Figure 8. Hepatic iron (A), nuclear factor erythroid 2-related factor 2 (NRF-2) (B), solute carrier family 7, member 11 (SLC7A-11) (C), and glutathione peroxidase (GPX-4) (D) in the different groups. SAE, Syzygium aromaticum extract; DOX, Doxorubicin. The values represent means ± S.D., n = 10. Means that do not share a letter showed a significant difference (p < 0.01).
Ijms 25 12541 g008
Figure 9. Hepatic ferritin heavy chain-1 (FTH-1) (A), acyl–CoA synthetase long-chain family member 4 (ACSL-4), (B), and nuclear receptor coactivator 4 (NCOA-4) (C) in different groups. The values represent means ± S.D. (n = 10). SAE, Syzygium aromaticum extract; DOX, Doxorubicin. Means that do not share a letter showed a significant difference (p < 0.05).
Figure 9. Hepatic ferritin heavy chain-1 (FTH-1) (A), acyl–CoA synthetase long-chain family member 4 (ACSL-4), (B), and nuclear receptor coactivator 4 (NCOA-4) (C) in different groups. The values represent means ± S.D. (n = 10). SAE, Syzygium aromaticum extract; DOX, Doxorubicin. Means that do not share a letter showed a significant difference (p < 0.05).
Ijms 25 12541 g009
Figure 10. (A) A photomicrograph of the liver section of the normal control group shows a normal hepatic structure, regular central veins (CVs), normal hepatocytes (Hs), normal blood sinusoids (Bs), and Kupffer cells (Ks). (B) Liver section of the SAE control group shows mostly normal hepatocytes, with normal Bs and Ks. (C) Liver section of the DOX-injected group exhibits disorganization of the hepatic architecture (dH), congested CVs (CCVs), cellular infiltrations, a vacuolated cytoplasm (V), pyknotic nuclei (arrows), irregular blood sinusoids (Bds), and distinct Ks. (D) Liver section of the DOX–SAE-treated group shows an improvement in the hepatic organization, represented by fewer congestions in the central vein (CV), fewer binucleated hepatocytes, and fewer cellular infiltrations. SAE, Syzygium aromaticum extract; DOX, Doxorubicin. (H&E × 400, scale bar = 50 μm).
Figure 10. (A) A photomicrograph of the liver section of the normal control group shows a normal hepatic structure, regular central veins (CVs), normal hepatocytes (Hs), normal blood sinusoids (Bs), and Kupffer cells (Ks). (B) Liver section of the SAE control group shows mostly normal hepatocytes, with normal Bs and Ks. (C) Liver section of the DOX-injected group exhibits disorganization of the hepatic architecture (dH), congested CVs (CCVs), cellular infiltrations, a vacuolated cytoplasm (V), pyknotic nuclei (arrows), irregular blood sinusoids (Bds), and distinct Ks. (D) Liver section of the DOX–SAE-treated group shows an improvement in the hepatic organization, represented by fewer congestions in the central vein (CV), fewer binucleated hepatocytes, and fewer cellular infiltrations. SAE, Syzygium aromaticum extract; DOX, Doxorubicin. (H&E × 400, scale bar = 50 μm).
Ijms 25 12541 g010
Table 1. Quantitative phytochemical analysis of S. aromaticum flower buds (SAFBs).
Table 1. Quantitative phytochemical analysis of S. aromaticum flower buds (SAFBs).
Phytochemical AnalysisSAFB
Total phenolic content (mg GAE/g DW)31.86 ± 2.43
Total flavonoid content (mg QE/g DW)18.65 ± 1.58
Total antioxidant capacity (TAC) (mg AAE/g DW)69.37 ± 3.79
Saponin (mg/g DW)365 ± 4.37
Anthocyanin (mg ECG/g DW)7.89 ± 0.45
DPPH scavenging activity (%)82.64% ± 3.95
IC50 of DPPH (mg/mL)6.05 ± 0.86 
SAFB, Syzygium aromaticum flower bud; GAE, Gallic acid equivalent; QE, Quercetin equivalents; DW, Dry weight; TAC, Total antioxidant capacity; AAE, Ascorbic acid equivalent; ECG, Epicatechin gallate; DPPH, Diphenyl-1-picrylhydrazyl; IC50, Half-maximal inhibitory concentration.
Table 2. GC-MS analysis of the phytochemical composition in SAE.
Table 2. GC-MS analysis of the phytochemical composition in SAE.
No.RT (min)NameMF.P.A (%)
17.64Ethyl-α-D-glucopyranosideC8H16O62.27
212.25EugenolC10H12O264.17
315.82CaryophylleneC15H2418.07
416.54HumuleneC15H243.83
519.50Caryophyllene oxideC15H24O0.86
621.5510-Heptadecen-8-ynoic acid, methyl esterC15H24O0.67
SAE, Syzygium aromaticum extract; RT, Retention time; MF, Molecular formula; P.A%, Peak area percentage.
Table 3. ADMET screening for each of DOX and Eugenol.
Table 3. ADMET screening for each of DOX and Eugenol.
Compound
(A1:S4)
DOXEugenol10-Heptadecen-8-ynoic Acid,
Methyl Ester
LogS−2.26−2.286−5.812
LogD0.4732.4184.205
LogP1.3752.2915.78
Pgp-inh0.0010.0030.389
Pgp-sub0.99800.002
HIA0.8290.0070.004
F (20%)0.0550.7320.208
F (30%)0.2090.9670.975
Caco-2−6.167−4.373−4.457
MDCK6.34 × 10−63.01 × 10−52.52 × 10−5
BBB0.0150.1880.713
PPB90.86%92.12%98.58%
VDss1.1770.8331.36
Fu10.82%3.22%0.83%
CYP1A2-inh0.2740.9010.924
CYP1A2-sub0.4950.9410.558
CYP2C19-inh0.0120.7160.882
CYP2C19-sub0.0640.6590.542
CYP2C9-inh0.0060.3130.708
CYP2C9-sub0.4060.8750.992
CYP2D6-inh0.0030.850.633
CYP2D6-sub0.1930.9210.641
CYP3A4-inh0.0880.2880.858
CYP3A4-sub0.1530.3710.129
CL13.02514.0424.467
T1/20.730.8870.379
hERG0.0250.0170.098
H-HT0.2610.0360.561
DILI0.9740.0460.91
Ames0.8290.0660.111
ROA0.0410.1210.026
FDAMDD0.3230.1530.024
Carcinogenicity0.680.4140.267
EC0.0030.7130.933
EI0.0120.9820.95
Respiratory0.8910.510.943
BCF0.5671.2032.424
IGC503.7343.6425.204
LC503.6533.9265.874
LC50DM5.554.7635.836
NR-AR0.0260.0890.363
NR-AR-LBD0.7930.0220.005
NR-AhR0.8660.4130.007
NR-Aromatase0.6440.020.442
NR-ER0.2170.2240.191
NR-ER-LBD0.5880.0360.016
NR-PPAR-gamma0.2210.0240.593
SR-ARE0.7710.140.336
SR-ATAD50.6420.2380.022
SR-HSE0.0270.0940.216
SR-MMP0.9450.1550.012
SR-p530.9790.0330.124
MW543.17164.08278.22
Vol516.727179.994326.919
Dense1.0510.9120.851
nHA1222
nHD910
TPSA212.3929.4626.3
nRot5312
nRing510
MaxRing1860
nHet1222
nRig2870
Flex0.1790.4294
nStereo500
Genotoxic Carcinogenicity Mutagenicity400
SureChEMBL101
Non-Biodegradable300
Skin Sensitization850
Toxicophores322
QED0.1470.6930.301
Synth5.0151.9612.503
Fsp30.370.20.722
MCE-18118.24360
Natural-Product-likeness1.371.0531.653
LipinskiRejectedAcceptedAccepted
PfizerAcceptedAcceptedRejected
GSKRejectedAcceptedAccepted
LogS, Log of the aqueous solubility; LogD, Log at physiological pH (7.4); LogP, Log of the octanol–water partition coefficient; Pgp-inh, P-glycoprotein inhibitors; Pgp-sub, P-glycoprotein substrates; HIA, human intestinal absorption; F (%), oral bioavailability; MDCK, Madin–Darby canine kidney; BBB, blood–brain barrier; PPB, plasma protein binding; VDss, volume of distribution; Fu, fraction unbound; CYP1A1, cytochrome P450 family 1 subfamily A; CL, clearance; T1/2, half-life time; hERG, human ether-à-go-go-related gene; H-HT, hereditary hemorrhagic telangiectasia; DILI, drug-induced liver injury; ROA, route of administration; FDAMDD, Food and Drug Administration maximum daily dose; EC, enteric coated; EI, enzyme induction; BCF, bioconcentration factor; IGC50, median inhibitory growth concentration; LC50, median lethal concentration; NR-AR-LBD, nuclear receptor–androgen receptor–ligand binding domain; AhR, aryl hydrocarbon receptor; ER, extended-release; SR, sustained-release; ATAD5, ATPase family A domain-containing protein 5; HSE, health, safety, and environment; MMP, matrix metalloproteinase inhibitor; MW, molecular weight; nHA, number of hydrogen bond acceptors; nHD, number of hydrogen bond donors; TPSA, topological polar surface area; nROT, number of rotatable bonds; nHet, number of heteroatoms; MaxRing, number of atoms in the biggest ring; nRing, number of rings; Flex, flexibility; nStereo, number of stereocenters; QED, quantitative estimate of drug-likeness; Fsp3, fraction of carbon atoms that are sp3 hybridized; MCE-18, medicinal chemistry evolution, 2018; GSK, GlaxoSmithKline.
Table 4. The ∆G and binding affinity (Kcal/mol).
Table 4. The ∆G and binding affinity (Kcal/mol).
CompoundGPX-4NRF-2SLC7A-11
Doxorubicin−6.7−7.8−8.4
10-Heptadecen-8-ynoic acid, methyl ester−3.9−4.3−5.3
Caryophyllene−5.2−6.1−7.3
Caryophyllene oxide−5.2−5.9−7.3
Ethyl_α_D-glucopyranoside−4.5−5.2−6.6
Eugenol−4.3−4.7−6.0
Humulene−5.3−5.9−7.4
Table 5. Serum alanine transaminase (ALT), aspartate transaminase (AST), alkaline phosphatase (ALP), total bilirubin (T.B), and direct bilirubin (D.B) in different groups.
Table 5. Serum alanine transaminase (ALT), aspartate transaminase (AST), alkaline phosphatase (ALP), total bilirubin (T.B), and direct bilirubin (D.B) in different groups.
GroupsALT (U/L)AST (U/L)ALP (U/L)T.B. (mg/dL)D.B. (mg/dL)
Negative control25.87 ± 0.76 e36.27 ± 1.21 c265.32 ± 4.67 a0.64 ± 0.013 a0.132 ± 0.004 c
SAE control23.44 ± 0.65 e32.44 ± 1.15 c258.79 ± 5.23 a0.58 ± 0.012 a0.126 ± 0.007 c
DOX-treated71.23 ± 1.78 a103.67 ± 2.78 e486.98 ± 6.34 b1.45 ± 0.034 b0.412 ± 0.008 a
DOX–SAE38.93 ± 0.85 d,e60.78 ± 2.56 d317.92 ± 5.79 e0.90 ± 0.016 a,c0.215 ± 0.006 d
The values represent the mean ± S.D., n = 10. SAE, Syzygium aromaticum extract; DOX, Doxorubicin. Means that do not share a letter in each column showed a significant difference (p < 0.05).
Table 6. The relative mRNA expression levels of the NRF2, SLC7A11, GPX4, FTH1, and NCOA4 genes of the different groups.
Table 6. The relative mRNA expression levels of the NRF2, SLC7A11, GPX4, FTH1, and NCOA4 genes of the different groups.
GroupsNRF2SLC7A11GPX4FTH1NCOA4
Negative control1.00 ± 0.00 c1.00 ± 0.00 a1.00 ± 0.00 e1.00 ± 0.00 b1.00 ± 0.00 a
SAE control1.56 ± 0.09 f1.12 ± 0.07 a1.39 ± 0.06 c1.07 ± 0.03 b1.02 ± 0.009 a
DOX-treated0.39 ± 0.04 a0.62 ± 0.08 b0.70 ± 0.05 b0.25 ± 0.06 a3.67 ± 0.12 b
DOX–SAE0.89 ± 0.07 c0.78 ± 0.06 a,b0.95 ± 0.08 e0.61 ± 0.08 c1.57 ± 0.08 a,c
The values represent the mean ± S.D., n = 10. SAE, Syzygium aromaticum extract; DOX, Doxorubicin. Means that do not share a letter in each column show a significant difference (p < 0.001).
Table 7. Hepatic inflammatory cytokines, including interleukin-6 (IL-6), interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), nuclear factor kappa-B (NF-κB), and cyclooxygenase-2 (COX-2), in the different groups.
Table 7. Hepatic inflammatory cytokines, including interleukin-6 (IL-6), interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), nuclear factor kappa-B (NF-κB), and cyclooxygenase-2 (COX-2), in the different groups.
GroupsIL-6
(Pg/mg Tissue)
IL-1β
(Pg/mg Tissue)
TNF-α
(Pg/mg Tissue)
NF-κB
(Pg/mg Tissue)
COX-2
(Pg/mg Tissue)
Negative control6.83 ± 0.56 e11.86 ± 1.15 a3.85 ± 0.29 c185.61 ± 4.23 a317.82 ± 6.48 c
SAE control6.12 ± 0.65 e12.08 ± 0.87 a3.27 ± 0.32 c170.85 ± 4.63 a296.73 ± 5.86 c
DOX-treated19.47 ± 0.93 a29.79 ± 1.85 c11.21 ± 1.04 b385.43 ± 6.24 b587.92 ± 7.58 a
DOX–SAE9.87 ± 0.76 d,e18.45 ± 1.06 e6.79 ± 0.75 e245.92 ± 5.85 c402.21 ± 6.46 d
The values represent the mean ± S.D., n = 10. SAE, Syzygium aromaticum extract; DOX, Doxorubicin. Means that do not share a letter in each column show significant differences (p < 0.001) or (p < 0.01).
Table 8. Effect of SAE treatment on the liver histopathologic score of the different groups.
Table 8. Effect of SAE treatment on the liver histopathologic score of the different groups.
GroupsHistopathologic Score
Negative control0.12 ± 0.09 a
SAE control0.10 ± 0.11 a
DOX-treated3.50 ± 0.21 c
DOX–SAE1.82 ± 0.19 b
Means that do not share a letter in each column show significant differences (p < 0.01).
Table 9. Forward and reverse primer sequences for RT-PCR.
Table 9. Forward and reverse primer sequences for RT-PCR.
GeneAccession NumberForward Sequence (5′–3′)Reverse Sequence (5′–3′)
NRF2NM_031789.3CACATCCAGACAGACACCAGTCTACAAATGGGAATGTCTCTGC
SLC7A11NM_001107673.3GAGGCGCTGTAGCCACATTAGGCATTCAACCAGGTGATCC
GPX4NM_008162CTCCATGCACGAATTCTCAGACGTCAGTTTTGCCTCATTG
FTH1NM_012848.2CCCTTTGCAACTTCGTCGCTCTCCGAGTCCTGGTGGTAGT
NCOA4NM_001034007.1TGAAGTGCAGTGCTCACACATTCGCTGCTGCTGACAGTTA
GAPDHNM_017008.4CCGCATCTTCTTGTGCAGTGGAGAAGGCAGCCCTGGTAAC
NRF2, Nuclear factor erythroid 2-related factor 2; SLC7A11, Solute carrier family 7, member 11; GPX4, Glutathione peroxidase; FTH1, Ferritin heavy chain-1; NCOA4, Nuclear receptor co-activator-4; GAPDH, Glyceraldehyde-3-phosphate dehydrogenase.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Alsirhani, A.M.; Abu-Almakarem, A.S.; Alwaili, M.A.; Aljohani, S.; Alali, I.; AlRashidi, A.A.; Abuzinadah, N.Y.; Alkhodair, S.A.; Mobasher, M.A.; Alothaim, T.; et al. Syzygium aromaticum Extract Mitigates Doxorubicin-Induced Hepatotoxicity in Male Rats. Int. J. Mol. Sci. 2024, 25, 12541. https://doi.org/10.3390/ijms252312541

AMA Style

Alsirhani AM, Abu-Almakarem AS, Alwaili MA, Aljohani S, Alali I, AlRashidi AA, Abuzinadah NY, Alkhodair SA, Mobasher MA, Alothaim T, et al. Syzygium aromaticum Extract Mitigates Doxorubicin-Induced Hepatotoxicity in Male Rats. International Journal of Molecular Sciences. 2024; 25(23):12541. https://doi.org/10.3390/ijms252312541

Chicago/Turabian Style

Alsirhani, Alaa Muqbil, Amal S. Abu-Almakarem, Maha Abdullah Alwaili, Salwa Aljohani, Ibtisam Alali, Aljazi Abdullah AlRashidi, Najlaa Yousef Abuzinadah, Sahar Abdulrahman Alkhodair, Maysa A. Mobasher, Tahiyat Alothaim, and et al. 2024. "Syzygium aromaticum Extract Mitigates Doxorubicin-Induced Hepatotoxicity in Male Rats" International Journal of Molecular Sciences 25, no. 23: 12541. https://doi.org/10.3390/ijms252312541

APA Style

Alsirhani, A. M., Abu-Almakarem, A. S., Alwaili, M. A., Aljohani, S., Alali, I., AlRashidi, A. A., Abuzinadah, N. Y., Alkhodair, S. A., Mobasher, M. A., Alothaim, T., Eid, T. M., & El-Said, K. S. (2024). Syzygium aromaticum Extract Mitigates Doxorubicin-Induced Hepatotoxicity in Male Rats. International Journal of Molecular Sciences, 25(23), 12541. https://doi.org/10.3390/ijms252312541

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop