Next Article in Journal
Noninvasive Methods to Detect Reactive Oxygen Species as a Proxy of Seed Quality
Next Article in Special Issue
Antioxidant Activity and Inhibition of Digestive Enzymes of New Strawberry Tree Fruit/Apple Smoothies
Previous Article in Journal
Repurposing Verapamil to Enhance Killing of T-ALL Cells by the mTOR Inhibitor Everolimus
Previous Article in Special Issue
Role of Dietary Polyphenols in the Activity and Expression of Nitric Oxide Synthases: A Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Rosemary (Rosmarinus officinalis L.) Glycolic Extract Protects Liver Mitochondria from Oxidative Damage and Prevents Acetaminophen-Induced Hepatotoxicity

by
Natalia S. S. Guimarães
1,
Vyctória S. Ramos
1,
Laura F. L. Prado-Souza
2,
Rayssa M. Lopes
2,
Gabriel S. Arini
3,
Luís G. P. Feitosa
3,
Ricardo R. Silva
3,
Iseli L. Nantes
2,
Debora C. Damasceno
4,
Norberto P. Lopes
3 and
Tiago Rodrigues
1,*
1
Interdisciplinary Center of Biochemistry Investigation, University of Mogi das Cruzes (UMC), Mogi das Cruzes CEP 08780-911, SP, Brazil
2
Center for Natural and Human Sciences, Federal University of ABC, Santo André CEP 09210-580, SP, Brazil
3
NPPNS, Department of Biomolecular Sciences, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto CEP 14040-900, SP, Brazil
4
Laboratory of Experimental Research on Gynecology and Obstetrics, Sao Paulo State University (UNESP), Botucatu CEP 18618-687, SP, Brazil
*
Author to whom correspondence should be addressed.
Submission received: 28 January 2023 / Revised: 26 February 2023 / Accepted: 28 February 2023 / Published: 3 March 2023

Abstract

:
Rosmarinus officinalis L. (rosemary) is an aromatic culinary herb. Native to the Mediterranean region, it is currently cultivated worldwide. In addition to its use as a condiment in food preparation and in teas, rosemary has been widely employed in folk medicine and cosmetics. Several beneficial effects have been described for rosemary, including antimicrobial and antioxidant activities. Here, we investigated the mechanisms accounting for the antioxidant activity of the glycolic extract of R. officinalis (Ro) in isolated rat liver mitochondria (RLM) under oxidative stress conditions. We also investigated its protective effect against acetaminophen-induced hepatotoxicity in vivo. A crude extract was obtained by fractionated percolation, using propylene glycol as a solvent due to its polarity and cosmeceutical compatibility. The quantification of substances with recognized antioxidant action revealed the presence of phenols and flavonoids. Dereplication studies carried out through LC-MS/MS and GC-MS, supported by The Global Natural Product Social Molecular Networking (GNPS) platform, annotated several phenolic compounds, confirming the previous observation. In accordance, Ro decreased the production of reactive oxygen species (ROS) elicited by Fe2+ or t-BOOH and inhibited the lipid peroxidation of mitochondrial membranes in a concentration-dependent manner in RLM. Such an effect was also observed in liposomes as membrane models. Ro also prevented the oxidation of mitochondrial protein thiol groups and reduced glutathione (GSH). In model systems, Ro exhibited a potent scavenger activity toward 2,2′-diphenyl-1-picrylhydrazyl (DPPH) radicals and superoxide anions. It also demonstrated an Fe2+ chelating activity. Moreover, Ro did not exhibit cytotoxicity or dissipate the mitochondrial membrane potential (∆Ψ) in rat liver fibroblasts (BRL3A cells). To evaluate whether such antioxidant protective activity observed in vitro could also be achieved in vivo, a well-established model of hepatotoxicity induced by acute exposure to acetaminophen (AAP) was used. This model depletes GSH and promotes oxidative-stress-mediated tissue damage. The treatment of rats with 0.05% Ro, administered intraperitoneally for four days, resulted in inhibition of AAP-induced lipid peroxidation of the liver and the prevention of hepatotoxicity, maintaining alanine and aspartate aminotransferase (ALT/AST) levels equal to those of the normal, non-treated rats. Together, these findings highlight the potent antioxidant activity of rosemary, which is able to protect mitochondria from oxidative damage in vitro, and effects such as the antioxidant and hepatoprotective effects observed in vivo.

1. Introduction

The Lamiaceae family contains a set of plants, often cultivated as herbs, whose leaves can be used as seasonings and whose essential oils are used as flavors in cosmeceuticals [1]. Among these plants, Rosmarinus officinalis L. stands out. Popularly known as rosemary (or alecrim in Brazil), R. officinalis is native to the Mediterranean region and is currently found worldwide. It has been extensively studied for its use in food and as a spice and/or preservative, in which it operates by inhibiting microbial growth and oxidative reactions [2]. The study of medicinal plants and herbs has proved to be an efficient strategy for prospecting novel compounds with therapeutic potential [3,4]. In this regard, a large number of secondary metabolites have been isolated and identified from Rosmarinus spp., including essential oils, flavonoids, tannins, terpenes, and phenolic acids [5,6,7]. In addition, carnosol, carnosic acid, and rosmarinic acids, which are reported to be the main components of rosemary, account for many of its biological activities [8,9,10]. The production of these bioactive secondary metabolites by rosemary depends on several factors, such as the plant part, climatic conditions and soil nutrients, humidity, temperature, water availability, and others [11]. Due to the large number of chemical studies carried out on R. officinalis, we opted for a dereplication strategy to avoid re-isolations [12]. Hyphenated liquid chromatography with electrospray ionization mass spectrometry (LC-ESI-MS/MS) is currently the most suitable technique for metabolomics approaches [13]. However, when analyzing extracts with an antioxidative potential, it is important to follow the oxidation processes at the source of the ESI in detail since substances with a low oxidation potential can lose electrons, changing the expected mass balance and leading to erroneous interpretations of chemical structures [14,15].
Furthermore, R. officinalis has long been used as a medicinal plant. In this context, a plethora of biological effects and health benefits have been attributed to rosemary essential oil, leaf extracts, and isolated substances, including antibacterial [16,17,18], antifungal [19,20], anti-inflammatory [21,22], antiatherogenic [23,24], antiangiogenic [25,26], antihypertensive [27], antiulcer [28], anti-diabetic [29,30], anticancer [31], and other effects (reviewed elsewhere [32,33]). In addition to these effects, this plant is also well known for its powerful antioxidant activity (reviewed in [34]). This is expected, as phytochemical analyses of rosemary extracts and essential oils have revealed several substances with recognized antioxidant properties, such as flavonoids and phenolic compounds [35,36]. In fact, such antioxidant activities of R. officinalis might help to explain some of the biological effects described above. The pathophysiology of several human diseases is related to the overproduction of reactive oxygen species (ROS) or a decreased capacity of the antioxidant defense system, resulting in oxidative stress, cellular/tissue damage, and, ultimately, organ dysfunction [37]. In this regard, it has been shown that R. officinalis exhibits hepatoprotective effects in different models of xenobiotic-induced liver toxicity, including carbon tetrachloride (CCl4) [38,39,40,41], creosote [38], azathioprine [39], hexavalent chromium [40], streptozotocin [41], and cyclophosphamide [42]. However, its protective effect against acetaminophen-induced hepatotoxicity has not been clearly demonstrated yet. Acetaminophen (AAP) is an anti-inflammatory and anti-pyretic drug, and AAP overdose is associated with acute liver failure [43]. During cytochrome P450 liver metabolism, AAP is transformed in the highly reactive metabolite N-acetyl-p-benzoquinone imine (NAPQI), which oxidizes GSH [44] and causes oxidative stress.
Although the antioxidant activity of R. officinalis has been extensively studied in several biological systems and toxicity models, its potential protective effect on mitochondria under oxidative stress conditions has not been investigated. Despite the central role they play in cellular metabolism and energy production, mitochondria are also often associated with oxidative status since superoxide radical anions (O2•−) are continuously generated during electron transport in the respiratory chain. This radical is counteracted by the antioxidant defense system, with the reducing power provided by reduced glutathione (GSH) and NAD(P)H [45]. However, excessive O2•− formation and, consequently, its conversion to hydrogen peroxide (H2O2), can produce the extremely reactive hydroxyl radical (OH) by means of the Fenton reactions, which are catalyzed by Fe2+ and other transition metals [46]. Thus, the investigation of the effects of R. officinalis in such a complex mitochondrial scenario can further elucidate the molecular mechanisms of antioxidant protection in biological systems. Here, we investigated the antioxidant properties of R. officinalis leaf extracts in rat liver mitochondria and addressed its protective effect against AAP-induced hepatotoxicity in rats.

2. Materials and Methods

2.1. Chemicals, Plant Source, and Extract Preparation

All reagents used in this study were of the highest commercially available grade of purity. Aqueous solutions were prepared using type I water obtained using a Milli-Q system (Millipore, USA). Rosmarinus officinalis L. species were collected in Brazil (GPS localization: 760 m, 23°29840′ S) during the winter (August) of 2018. After botanical identification, voucher specimens were deposited at the Herbarium Mogiense (University of Mogi das Cruzes, Brazil). After drying at 40 °C in a ventilated drying oven, 100 g of selected leaves were submitted to fractionated percolation using a mixture of propylene glycol/water (7/3) as solvent, resulting in 100 mL of Rosmarinus officinalis glycolic extract (Ro). This extract was considered a 100% crude extract for subsequent concentration calculations.

2.2. Isolation of Mitochondria, Animal Treatments, and Sample Preparation

Liver mitochondria were isolated from rats by differential centrifugation in isosmotic media, as previously described [47]. Adult male Wistar rats weighing approximately 180 g were randomly divided into four groups (n = 7), kept in a 12/12 h light/dark cycle, and fed ad libitum. All experiments involving animals were conducted in accordance with the guide for the care and use of laboratory animals (NIH Guidelines) and were previously approved by the Animal Ethics Committee of University of Mogi das Cruzes (CEUA). In order to induce hepatotoxicity, the AAP group received a single dose of 900 mg/kg of acetaminophen (Sigma-Aldrich, St. Louis, MI, USA) intraperitoneally. Animals were sacrificed 4 h after drug administration. The control group (C) received the same volume of solvent used for AAP dissolution, i.e., dimethyl sulfoxide (DMSO; Sigma-Aldrich, USA). The other two groups received 0.1 mL of 0.5% Ro intraperitoneally once per day in the morning for four consecutive days. One of these groups (Ro/AAP) also received AAP on the last day, as described above, 4 h before sacrifice. The animals were anesthetized with xylazine (5 mg/kg) and ketamine (60 mg/kg) for cardiac blood collection. The blood was immediately centrifuged at 700× g for 5 min to achieve serum separation. For homogenate preparation, the livers were removed and cut into small fragments in a buffer containing 250 mmol/L sucrose, 1 mmol/L EGTA, and HEPES- KOH 10 mmol/L at a pH of 7.4 at 4 °C. After washing the samples twice with the same medium, the tissues were homogenized using a Potter-Elvehjem. This was followed by centrifugation at 580× g for 5 min at 4 °C. The supernatant, called a homogenate, was immediately frozen at −70 °C for subsequent experiments.

2.3. Preparation of PCPECL Liposomes

The phospholipids phosphatidylcholine, phosphatidylethanolamine, and cardiolipin (PCPECL, 5:3:2 ratio) were dissolved in CHCl3, which was further evaporated under argon flux. The lipidic film was hydrated with 10 mM of cold sodium phosphate buffer at a pH of 7.4 and vortexed. To obtain the liposome solution (1 mM), lipids were submitted to sonication for 30 min at 4 °C using a Ney Ultrasonik (J. M. Ney Co., Bloomfield, CT, USA).

2.4. Total Phenols and Flavonoids

The soluble phenol derivatives were estimated using the Folin–Ciocalteu method [48] and expressed as μM of gallic acid equivalents based on a standard curve [49]. For flavonoid quantification, an aliquot was incubated in a medium containing 60 μL of glacial acetic acid, 1.0 mL of pyridine H2O:AlCl3 12% solution (17:80:3), and 1.24 mL of DMSO:H2O (1:1) for 5 min at 25 °C. The absorbance of the chromophore produced in this reaction was determined spectrophotometrically at 420 nm, and the flavonoid content was determined based on a standard curve and expressed as µM of quercetin equivalents.

2.5. DPPH Assay, Fe2+, and Superoxide Scavenger Activity

The reduction of the 1,1′-diphenyl-2-picrylhydrazyl radical (DPPH, Sigma-Aldrich, USA) by Ro was accompanied by photometric measurement in a UV1800 spectrophotometer (Shimadzu, Kyoto, Japan). An amount of 1.5 mL of 40 mM sodium acetate pH 5.5 was mixed into the reaction with 1.0 mL of absolute ethanol containing DPPH to achieve a final concentration of 0.1 mM. After incubation with different Ro concentrations for 5 min at 25 °C, the final absorbance was measured at 517 nm. Quercetin (Q, Sigma-Aldrich, USA) was used as reference. The amount of Fe2+ was quantified photometrically at 535 nm using 0.2 mM bathophenanthroline disulfonic acid (Sigma-Aldrich, USA) [50] in a competitive assay with Ro. At last, the xanthine/xanthine oxidase system was used to generate O2•−. The scavenger activity of Ro was estimated by the inhibition of the nitroblue tetrazolium (NBT, Sigma-Aldrich, USA) reduction. After adding 0.08 U/mL xanthine oxidase to a phosphate buffer at a pH of 7.5, which contained 0.05 mM EDTA, 0.2 mM hypoxanthine, and 0.1 mM NBT, the absorbance was measured at 540 nm after 20 min of incubation at 37 °C (Shimadzu UV1800 Spectrophotometer, Tokyo, Japan).

2.6. Reactive Oxygen Species (ROS)

The production of mitochondrial ROS was estimated kinetically using 2′,7′-dichlorodihydrofluorescein diacetate (H2DCFDA, Sigma-Aldrich, USA) [51], as previously described [52]. Briefly, mitochondria (1 mg/mL) were incubated in a buffer containing 125 mM sucrose, 65 mM KCl, and 10 mM HEPES-KOH at a pH of 7.4, plus 1 µM H2DCFDA at 30 °C, under continuous stirring in the presence of 5 mM potassium succinate (Sigma-Aldrich, USA) (plus 2 μM rotenone, Sigma-Aldrich, USA). The fluorescence emissions were recorded in a Hitachi F-2500 Spectrophotometer (Tokyo, Japan) at 503/529 nm for excitation/emission wavelength pairs, respectively.

2.7. Lipid Oxidation

The liposomes (1 mM), mitochondria (1 mg/mL), or liver homogenates (1 mg/mL) were incubated with or without different Ro concentrations at 37 °C for 30 min, and with 50 μM Fe(NH4)2(SO)4 (plus 2.0 mM sodium citrate) or 0.6 mM t-BOOH as inducers of oxidative stress. Following incubation, for a thiobarbituric acid reactive substances (TBARS) assay, 1% thiobarbituric acid (TBA, Sigma-Aldrich, USA) was prepared by adding 50 mM NaOH, 15 μL of 10 M NaOH, and 75 μL of 20% H3PO4 to each sample, followed by further incubation for 20 min at 85 °C. The MDA-TBA complex was extracted with 2 mL of n-butanol, and the absorbance was measured at 535 nm. The TBARS were calculated from ε = 1.56 × 105 M−1.cm−1, as described in [53]. Lipid hydroperoxides (LOOH) were also quantified in isolated mitochondria using xylenol orange, as previously described in [54]. When applied, the percentages of inhibition by Ro were calculated in relation to positive controls (t-BOOH or Fe2+) which were considered to achieve 100% inhibition.

2.8. GSH and Protein Thiol Groups

After 30 min of incubation at 37 °C in a medium containing 125 mM sucrose, 65 mM KCl, and 10 mM HEPES-KOH at a pH of 7.4, plus 5.0 mM potassium succinate and 2.0 μM rotenone, the mitochondrial suspensions or liver homogenates were treated with 0.5 mL of 13% trichloroacetic acid and centrifuged at 400× g for 5 min. For the GSH assay, aliquots (100 µL) of the supernatant were mixed with 1.8 mL of a 0.1 M sodium phosphate buffer at a pH of 8.0 containing 5 mM EGTA plus 100 μL of 1 mg/mL o-phthalaldehyde (Sigma-Aldrich, USA). After 15 min of incubation, the fluorescence emissions were detected at 350/420 nm (excitation/emission) in a Hitachi F-2500 (Japan) fluorescence spectrophotometer [55]. The reduced thiol groups of proteins were quantified using 5,5′-dithiobis(2-nitrobenzoic acid) (DTNB, Sigma-Aldrich, USA) [56]. The pellet obtained from the acid precipitation described above was suspended with 1 mL of a 0.5 M potassium phosphate buffer at a pH of 7.6 which contained 0.7% SDS. After adding 100 μM DTNB, the absorbance was measured at 412 nm and the amount of the reduced thiol groups was calculated from ε = 13,600 M−1.cm−1 [57].

2.9. Cell Culture and Cellular Assays

BRL 3A cells (CRL-1442, ATCC) were cultivated in high-glucose Dulbecco’s modified Eagle’s medium (DMEM; Sigma-Aldrich, USA) supplemented with 10% fetal bovine serum (FBS) (Gibco, Thermo Fischer Scientific, Waltham, MA, USA), 100 U/mL penicillin, and 100 μg/mL streptomycin at 37 °C in a 5% CO2 atmosphere (Panasonic MCO-19AIC incubator, Osaka, Japan). Cells were used for the experiments during 4–8 passages after thawing. For the experiments, cells were washed twice with a calcium- and magnesium-free buffered saline solution (CMF-BSS), detached from the flasks using trypsin/EDTA (Gibco, Thermo Fischer Scientific, USA), and suspended in the supplemented medium. For the viability assay, cells (5 × 104 cells/cm2) were added to 96-well microplates (0.2 mL final volume) and incubated with different concentrations of Ro for 24 h. After adding 0.25 mg/mL MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide] (Sigma-Aldrich, USA), 4 h of incubation, and the solubilization of formazan crystals, the absorbance of each well was measured at 570 nm in a Biotek ELX 800 microplate reader (BioTek Instruments, Winooski, VT, USA). Cell viability was determined relative to the control (absence of Ro), which was considered to have a 100% viability [58]. The mitochondrial transmembrane potential in the BRL 3A cells was estimated using fluorescence microscopy. Cells (5 × 104 cells/cm2) were seeded in 3.5 cm glass dishes (with a 0.17 mm thick cover glass on the bottom (Greiner Bio-One, Frickenhausen, Germany) and loaded with 40 nM DiOC6(3) (Thermo Fisher Scientific, USA) and 5 nM Hoechst 33258 (Thermo Fisher Scientific, USA) with incubation at 37 °C for 30 min. Images were acquired in a widefield Leica AF6000 microscope (Leica Microsystems, Wetzlar, Germany), using the HCX APO UVI 100×/1.3 oil plan apochromatic objective and the ultrafast Leica DFC365 FX monochromatic digital camera (Leica Microsystems, Germany). The set of cube filters used included A4 (ex 360/40, DC 400 nm, em 470/40) and L5 (ex 480/40, DC 505, em 527/30) (Leica Microsystems, Germany) filters.

2.10. Blood Biochemical Analyses

Aspartate aminotransferase (AST) and alanine aminotransferase (ALT) were determined in serum using commercial kits according to the manufacturer’s instructions (LABTEST, Lagoa Santa, MG, Brazil).

2.11. Untargeted Metabolomics Analysis by HPLC-ESI-MS/MS

An HPLC-MS analysis was performed in a Shimadzu UFLC system coupled to a quadrupole time-of-flight mass spectrometer (micrOTOF QII, Bruker Daltonics, Billerica, MA, USA) using a C18 column (5 µm, XB-C18 Kinetex, 100Å, 150 × 3 mm, Phenomenex). The mobile phase was composed of water (A) and MeOH (B), both with 0.1% formic acid, at a flow rate of 0.75 mL.min−1. The following gradient was employed: 0–23 min, 10–100% B; 23–26 min, 100% B; 26–27 min, 100–10% B; and 27–30 min, 10% B. The column oven was set to 45 °C, and an injection volume of 10 µL was selected. Chromatograms were acquired in both positive and negative ionization modes, and the following parameters were employed for the mass spectrometer: end plate offset, 500 V; capillary voltage, 3200 V for negative mode and 3500 V for positive mode; nebulizer pressure, 4.5 bar; dry gas (N2) flow, 9 L.min−1; dry temperature, 200 °C; mass range, m/z 150 to 1200; MS/MS scan mode; number of precursors, 3; exclusion activation, 1 spectrum; and exclusion release, 36 s.

2.12. Gas Chromatography-Mass Spectrometry (GC-MS) Analysis

The glycolic extract was submitted to a head space analysis, and the GC-MS methodology was based on the previously reported methodology [59]. The analysis was carried out on the GCMS-QP2010 (Schimadzu, Japan) according to the following parameters: injector temperature, 250 °C; column temperature, 60 °C; heating ramp from 60 to 210 °C, at 3 °C/min, with a total time of 50 min; chromatographic column, DB-5, 30 m × 0.25 mm in diameter, 0.25 μm in thickness; and helium was used as the carrier gas under 79.7 kPa at 1.30 mL/min with a linear velocity of 41.6 cm/s, a 1 μL injection volume, and a 1:60 split.

2.13. Statistical Analyses

Quantitative data are presented as the mean ± SD of (at least) three independent experiments performed in triplicate. For multiple comparisons, a one-way ANOVA was used, followed by Tukey’s post hoc test. The Prism 8.0 software (GraphPad Software Inc., La Jolla, CA, USA) was used to perform the data analyses. Statistical significance was defined as * (p < 0.05), ** (p < 0.01), and *** (p < 0.001).

3. Results

3.1. Rosmarinus officinalis Glycolic Extract (Ro) Protects Mitochondrial Lipids and Proteins from Oxidation

The production of mitochondrial ROS was evaluated through the kinetic measurement of dichlorofluorescein (DCF) fluorescence. When incubated with t-BOOH or Fe2+, isolated rat liver mitochondria energized by succinate immediately increased the production of ROS. Due to the nature of the oxidative stress inducer, it was possible to observe that the fluorescence increase rate was faster for the Fe2+ (Figure 1B, black line) than for the t-BOOH (Figure 1A, red line). The preincubation of the mitochondrial suspension with Ro abolished the ROS production triggered by both inducers in a concentration-dependent manner. These effects were translated into the protection of mitochondrial membranes from oxidation by Fe2+ (squares, orange) or t-BOOH (circles, red), estimated by TBARS (Figure 1C). Moreover, the formation of Fe2+-induced lipid hydroperoxides was inhibited by Ro at 0.025% (orange) and 0.05% (red) (Figure 1D). Such a lipid protective action exhibited by Ro was accompanied by the prevention of GSH oxidation by t-BOOH (Figure 1E) and the oxidation of reduced thiol groups of mitochondrial proteins by both t-BOOH (Figure 1F) and Fe2+ (Figure 1G) at the same concentrations.
Together, these results indicate the ability of Ro to protect mitochondria (and possibly cells) from oxidative damage, highlighting its potential to prevent pathological conditions and diseases associated with oxidative stress.

3.2. Iron (II) Chelating and Free Radical Scavenger Activity Account to the Antioxidant Protection Exhibited by Ro

The quantification of total phenols and flavonoids, substances with recognized antioxidant action, are presented in Table 1. In an attempt to provide further mechanistical insights regarding the antioxidant capacity of Ro, its ability to chelate Fe2+, which is used here as inducer of oxidative stress, was investigated. In a competitive spectrophotometric assay using bathophenanthroline, Ro chelated more than 75% of the available Fe2+, even at a lower concentration (Figure 2A). This may help to explain, at least partially, the protective effects observed in the Figure 1. Additionally, the free radical scavenger activity of Ro was investigated. As observed in Figure 2B, Ro reduced (scavenged) DPPH radicals in a concentration-dependent manner. At 0.025%, the effect was similar to 10 μM quercetin, a well-studied flavonoid known for its free radical scavenger activity and antioxidant properties [60]. Since DPPH is not a biological free radical, we also investigated the ability of Ro to scavenge O2•− generated by the xanthine/xanthine oxidase system, using NBT as indicator [61]. Ro exhibited significant O2•− scavenger activity at 0.05 and 0.01%, evaluated by the inhibition of NBT reduction by this radical. Finally, using a lipidic model system to mimic mitochondrial membranes [62], i.e., unsaturated PCPECL liposomes, it was possible to demonstrate that Ro prevents Fe2+-induced lipid oxidation regardless of mitochondrial function or a dependence on mitochondrial constituents. Thus, the ability to scavenge free radicals and to block lipid peroxidation reactions contributed to Ro’s antioxidant activity and to the protective effects observed in mitochondria (Figure 1).

3.3. Ro Does Not Affect Viability or Mitochondrial Membrane Potential (∆Ψ) of Liver Fibroblasts

To strengthen the pharmacological potential of Ro, its cytotoxicity was evaluated in vitro. This is relevant since many antioxidant substances also have a cytotoxic effect [63] which can hinder their therapeutic potential. In this regard, the possible cytotoxicity of Ro was investigated in cultured BRL 3A cells, a fibroblast-like cell line isolated from rat livers. Cell viability, evaluated by the MTT assay, was not affected by Ro in the concentration range tested (0.005–0.05%), i.e., the incubation of Ro with BRL3A cells for 24 h did not decrease cell viability compared to the control (Figure 3A). These results were confirmed by the Trypan blue exclusion assay (Figure 3B). Moreover, Ro did not alter the ∆Ψ of BRL 3A cells assessed by fluorescence microscopy using DiOC6(3) (Figure 3C(b),D, red bar). As a control, the total ∆Ψ dissipation was achieved by adding CCCP, an uncoupler of the OXPHOS (Figure 3C(c),D, white bar).

3.4. Ro Protection against Acetaminophen-Induced Hepatotoxicity in Rats

The antioxidant activity of Ro, its effect in protecting isolated rat liver mitochondria from oxidative stress conditions, and its absence of cytotoxicity in vitro led us to investigate whether such protective effects can be observed in vivo. To this end, a well-established and clinically relevant model of oxidative-stress-mediated hepatotoxicity was selected. The acute exposure to high doses of AAP generates radical intermediates during its hepatic metabolism, depleting antioxidant defenses and resulting in oxidative tissue damage [43]. To establish the experimental model, a single dose of AAP (900 mg/kg) was administrated to the rats, which were or were not treated with 0.5% Ro, and the blood levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were analyzed. This high dose of AAP is well established in the literature to induce acute hepatotoxicity in mice and rats [64,65]. As expected, AAP administration increased serum levels of ALT (Figure 4A, black bar) and AST (Figure 4B, black bar), indicating the occurrence of liver injury. As a control, Ro alone did not exert any effect on ALT or AST levels (orange bars, Figure 4A,B, respectively) compared to the basal levels, i.e., control, non-treated rats (white bars, Figure 4A,B), indicating the absence of an intrinsic hepatotoxicity of the extract.
Such a hepatotoxic effect of AAP was accompanied by GSH consumption (Figure 4C, black bar) and increased lipid oxidation (Figure 4D, black bar) without affecting the reduced thiol levels of proteins (Figure 4E, black bar). Interestingly, the treatment of animals with 0.5% Ro was able to reestablish the basal levels of ALT and AST in AAP-treated animals (red bars, Figure 4A,B, respectively), indicating a complete restoration of the liver integrity against the AAP-induced toxicity. We then further investigated whether this effect was accompanied by an improvement in oxidative stress indicators. In AAP-treated rats, Ro partially inhibited GSH oxidation (Figure 4C, red bar) and completely prevented lipid oxidation (Figure 4D, red bar) without an effect on the redox state of the protein thiol groups (Figure 4E, red bar). It is noteworthy that Ro diminished even the basal lipid oxidation (Figure 4D, orange bar).

3.5. Chemical Composition of Rosmarinus officinalis L. Glycolic Extract

In order to gain more information about the chemical composition of Ro, we conducted an untargeted metabolomics analysis of the glycolic extract at both positive and negative ionization modes. The annotated metabolites are shown in Figure 5/Table 2 and the ion intensity of each metabolite are presented in Figure 6. As expected, several phenolic compounds already described for this plant were detected [36], e.g., caffeic acid, rosmanol, and rosmarinic and carnosinic acids. All metabolites had their annotations confirmed through a detailed discussion of the decomposition reactions in the gaseous phase [66], following the correlation with the literature recently proposed by Pilon and collaborators [67] for the analysis of glycosylated flavonoids.

4. Discussion

The wide range of biological actions reported for R. officinalis has long drawn the attention of researchers worldwide. In this study, for the first time, we showed that the antioxidant activity exhibited by rosemary extract and its chemical constituents is able to protect mitochondria subject to oxidative stress conditions. During the electron transport by the respiratory chain, mitochondria generate superoxide anions that are converted to hydrogen peroxide (H2O2). In the presence of Fe2+, H2O2 is converted to hydroxyl radicals, and these species are able to oxidize mitochondrial lipids and proteins, compromising energy production and Ca2+ homeostasis and culminating in cell death [46,72]. Thus, we used this complex system to evaluate the antioxidant potential of the glycolic extract of R. officinalis (Ro). Two different methods of inducing oxidative stress were used: Fe2+ and t-butyl hydroperoxide (t-BOOH). The former is able to catalyze Fenton reactions, amplifying the production of reactive oxygen species (ROS). and the latter is an organic peroxide that per se consumes antioxidant defenses to be eliminated, exposing cells to oxidative damage [45,46]. In both situations, Ro was able to inhibit ROS production and the oxidation of mitochondrial lipids and proteins. Previous studies have reported the ability of R. officinalis to inhibit the lipid peroxidation of phospholipids and membrane model systems [73,74,75], erythrocytes [76,77], low density lipoproteins (LDL) [78], and tissue homogenates [28].
Among the mechanisms of action for antioxidant activity, we found that Ro exhibits a free radical scavenger activity and the ability to chelate Fe2+. Most studies have used a DPPH assay to infer the scavenger activity of rosemary extracts and essential oils; however, it is noteworthy that DPPH is not a radical produced in biological systems. Therefore, we further examined and demonstrated the capacity of the rosemary leaf glycolic extract to scavenge superoxide anions. Such an ability was previously shown for diterpenoids isolated from R. officinalis [79], and it is subject to seasonal variations [80]. The essential oil of R. officinalis was also able to scavenge hydroxyl radicals and chelate Fe2+ [81]. These properties of R. officinalis extracts are provided by their high content of powerful, well-known, synergistically acting antioxidant compounds including flavonoids, phenolic acids, and terpenes. For example, the antioxidant activity exhibited by carnosol was linked to an enhanced health and lifespan in C. elegans, accompanied by an increase in the activity of several antioxidant enzymes and a decrease in TBARS content [82]. As expected, the phytochemical analysis of the glycolic extract of R. officinalis identified the presence of several phenolic compounds already described for this plant. It should be noted that chlorogenic acid, a common compound with significant antioxidant activity, was not observed in these samples. A recent study demonstrated that the concentration of chlorogenic acid varies greatly depending on the drying process used for the leaves [83], which may explain, in part, the absence of the signal. The polyphenolic profile of rosemary has been widely described in the scientific literature [70,84,85,86], and the profile observed in the glycolic extract of the R. officinalis employed in this study was characterized by the presence of carnosic acid, carnosol, rosmarinic acid, and hesperidin. The mechanism of action was related to the free radicals’ chain terminators and scavengers of ROS. On the other hand, compounds such as hesperidin are known for their ability to chelate metals. This confers a resistance to pests, as described in citrus [87], but can also support antioxidant properties. The occurrence of astragalin reinforces all the biological effects described in our article. This kaempferol 3-glucoside has several biological activities described, including antioxidant effects [88]. Therefore, the global analysis of the main metabolites contained in the extract support the observed activities. Finally, as expected, GC-MS exhibited a trace signal. The major compound detected was camphor, showing no contribution of the previous essential oil constituents to the described biological activities.
The antioxidant power of R. officinalis has been shown to be responsible for the plant’s protective effects against oxidative stress. Rosemary prevented the renal toxicity elicited by diethylnitrosamine [89] and potassium dichromate [90] by inhibiting lipid peroxidation and improving the capacity of the antioxidant enzymatic system. Potassium dichromate was also employed to induce hepatotoxicity in rats. In this context, rosemary essential oil exhibited hepatoprotective action, characterized by the inhibition of the lipid peroxidation, GSH, and protein oxidation, and by the restoration of the levels of antioxidant enzymes [40]. Similar observations were made for hepatotoxicity induced by CCl4. Rosemary essential oil inhibited the lipid peroxidation and ‘reversed’ the activities of antioxidant enzymes catalase, peroxidase, glutathione peroxidase, and glutathione reductase in a liver homogenate [91]. A methanolic extract obtained from R. officinalis leaves [92,93] and a shoot tincture [94] also presented the same protective results on CCl4-induced hepatotoxicity in rats. When t-BOOH was used as the oxidizing agent used to induce liver damage, R. officinalis also exhibited protective effects [95]. Acetaminophen is one of the most widely used analgesic and antipyretic drugs in the world. Although it is a relatively safe drug at therapeutic doses, acute overdose, chronical exposition, or association with other xenobiotics can result in severe hepatotoxicity. After oral intake, a major part of it is conjugated with glucuronic acid in the liver and eliminated by the kidneys. However, a fraction of AAP undergoes metabolization by the cytochrome P450 system, generating the toxic metabolite N-acetyl-p-benzoquinone imine (NAPQI) which, under normal conditions, is conjugated with GSH and eliminated. Nonetheless, increased NAPQI production results in the depletion of GSH and, consequently, oxidative liver damage [96]. Thus, we investigated for the first time the possible protective effect of Ro on AAP-induced hepatotoxicity. To validate the model, our data showed that a single high dose of AAP caused a depletion of GSH and increased lipid peroxidation. Such oxidative alterations were accompanied with liver damage. This was attested by increased levels of ALT and AST, biochemical markers of hepatocyte damage. Both aminotransferases were quantified since tissues other than the liver have elevated AST levels, such as the erythrocytes, heart, and muscle, while ALT has more specificity to the liver. Our data showed an increased AST/ALT ratio (De Ritis ratio), higher than 1.0, when rats were treated with AAP. This is predictive of long-term complications such as fibrosis and cirrhosis [97]. As shown, Ro indisputably protected the rat livers from AAP toxicity since the AST and ALT levels returned to the basal levels when compared to control. The powerful antioxidant activity of Ro seems to be responsible for this hepatoprotection since the GSH depletion was abolished and the lipid oxidation was also diminished. This strategy of using antioxidants to protect the liver from AAP toxicity was successfully achieved with N-acetyl cysteine [98,99].
Finally, it is important to correlate the general chemistry data annotated in this paper with the Ro effects. More than half of the compounds have the catechol group. It is well known that catechol and its many functionalized derivatives are excellent metal chelators [100]. Considering only the phenol group, which is a good and well-known antioxidant [100], the number is even more expressive since all 18 major compounds noted have at least one hydroxyl linked to an aromatic ring. Therefore, we believe that the observed effect must surely be a synergistic combination of all the actives described in this work.

5. Conclusions

Together, our data reinforced the antioxidant activity of R. officinalis through a free radical scavenger activity and by diminishing the Fe2+-catalyzed Fenton reactions. We showed that these activities enabled Ro to protect isolated rat liver mitochondria from oxidative damage caused by different mechanisms (Fe2+ or t-BOOH). Moreover, Ro did not present cytotoxicity in cultured liver cells, and it exhibited a potent hepatoprotective effect against AAP toxicity in vivo. Further studies are necessary to the development of pharmaceutical formulations containing Ro, and clinical trials are required to evaluate the potential hepatoprotective effect in patients.

Author Contributions

Conceptualization, T.R.; formal analysis, T.R. and N.P.L.; investigation, N.S.S.G., V.S.R., L.F.L.P.-S., R.M.L., G.S.A., L.G.P.F. and R.R.S.; resources, I.L.N., D.C.D. and T.R.; data curation, N.S.S.G. and T.R.; writing—original draft preparation, N.P.L. and T.R.; writing—review and editing, I.L.N., D.C.D., N.P.L. and T.R.; visualization, N.S.S.G. and V.S.R.; supervision, T.R.; project administration, T.R.; funding acquisition, T.R. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP, 2008/01724-4; 2018/25747-5; 2020/02207-5; 2021/14650-3), the Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES, Financial Code 001), and the Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq, 312020/2019-8).

Institutional Review Board Statement

The study was conducted according to the guide for the care and use of laboratory animals (NIH guidelines), the Guiding Principles for Biomedical Research Involving Animals from the National Council for Animal Experimentation (CONCEA, Brazil) and approved by the Institutional Animal Care and Use Committee at the University of Mogi das Cruzes CEUA/UMC.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank Carla T. Graves and Thiago E. R. dos Santos for the technical assistance with the animal manipulation and treatments. We also thank Marilia Elias Gallon and Izabel Cristina Casanova Turatti for the technical assistance with the mass spectrometry experiments.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ramos Da Silva, L.R.; Ferreira, O.O.; Cruz, J.N.; De Jesus Pereira Franco, C.; Oliveira Dos Anjos, T.; Cascaes, M.M.; Almeida Da Costa, W.; Helena De Aguiar Andrade, E.; Santana De Oliveira, M. Lamiaceae Essential Oils, Phytochemical Profile, Antioxidant, and Biological Activities. Evid.-Based Complement. Altern. Med. 2021, 2021, 6748052. [Google Scholar] [CrossRef]
  2. Veenstra, J.P.; Johnson, J.J. Rosemary (Salvia rosmarinus): Health-Promoting Benefits and Food Preservative Properties. Int. J. Nutr. 2021, 6, 1. [Google Scholar] [CrossRef] [PubMed]
  3. Gurib-Fakim, A. Medicinal Plants: Traditions of Yesterday and Drugs of Tomorrow. Mol. Asp. Med. 2006, 27, 1–93. [Google Scholar] [CrossRef] [PubMed]
  4. Boldi, A.M. Libraries from Natural Product-like Scaffolds. Curr. Opin. Chem. Biol. 2004, 8, 281–286. [Google Scholar] [CrossRef]
  5. Bouyahya, A.; Et-Touys, A.; Bakri, Y.; Talbaui, A.; Fellah, H.; Abrini, J.; Dakka, N. Chemical Composition of Mentha pulegium and Rosmarinus officinalis Essential Oils and Their Antileishmanial, Antibacterial and Antioxidant Activities. Microb. Pathog. 2017, 111, 41–49. [Google Scholar] [CrossRef] [PubMed]
  6. Mahmoud, A.A.; Al-Shihry, S.S.; Son, B.W. Diterpenoid Quinones from Rosemary (Rosmarinus officinalis L.). Phytochemistry 2005, 66, 1685–1690. [Google Scholar] [CrossRef]
  7. Borges, R.S.; Ortiz, B.L.S.; Pereira, A.C.M.; Keita, H.; Carvalho, J.C.T. Rosmarinus officinalis Essential Oil: A Review of Its Phytochemistry, Anti-Inflammatory Activity, and Mechanisms of Action Involved. J. Ethnopharmacol. 2019, 229, 29–45. [Google Scholar] [CrossRef] [PubMed]
  8. Bai, N.; He, K.; Roller, M.; Lai, C.S.; Shao, X.; Pan, M.H.; Ho, C.T. Flavonoids and Phenolic Compounds from Rosmarinus officinalis. J. Agric. Food Chem. 2010, 58, 5363–5367. [Google Scholar] [CrossRef]
  9. Jordán, M.J.; Castillo, J.; Bañón, S.; Martínez-Conesa, C.; Sotomayor, J.A. Relevance of the Carnosic Acid/Carnosol Ratio for the Level of Rosemary Diterpene Transfer and for Improving Lamb Meat Antioxidant Status. Food Chem. 2014, 151, 212–218. [Google Scholar] [CrossRef]
  10. Ibrahim, N.; Abbas, H.; El-Sayed, N.S.; Gad, H.A. Rosmarinus officinalis L. Hexane Extract: Phytochemical Analysis, Nanoencapsulation, and in Silico, in Vitro, and in Vivo Anti-Photoaging Potential Evaluation. Sci. Rep. 2022, 12, 13102. [Google Scholar] [CrossRef]
  11. Borrás-Linares, I.; Stojanović, Z.; Quirantes-Piné, R.; Arráez-Román, D.; Švarc-Gajić, J.; Fernández-Gutiérrez, A.; Segura-Carretero, A. Rosmarinus officinalis Leaves as a Natural Source of Bioactive Compounds. Int. J. Mol. Sci. 2014, 15, 20585–20606. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. De Oliveira, G.G.; Carnevale Neto, F.; Demarque, D.P.; de Sousa Pereira-Junior, J.A.; Sampaio Peixoto Filho, R.C.; de Melo, S.J.; da Silva Almeida, J.R.G.; Lopes, J.L.C.; Lopes, N.P. Dereplication of Flavonoid Glycoconjugates from Adenocalymma imperatoris-maximilianii by Untargeted Tandem Mass Spectrometry-Based Molecular Networking. Planta Med. 2017, 83, 636–646. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Demarque, D.P.; Dusi, R.G.; de Sousa, F.D.M.; Grossi, S.M.; Silvério, M.R.S.; Lopes, N.P.; Espindola, L.S. Mass spectrometry-based metabolomics approach in the isolation of bioactive natural products. Sci Rep. 2020, 10, 1051. [Google Scholar] [CrossRef] [Green Version]
  14. Guaratini, T.; Vessecchi, R.L.; Lavarda, F.C.; Campos, P.M.B.G.M.; Naal, Z.; Gates, P.J.; Lopes, N.P. New chemical evidence for the ability to generate radical molecular ions of polyenes from ESI and HR-MALDI mass spectrometry. Analyst 2004, 129, 1223–1226. [Google Scholar] [CrossRef]
  15. Guaratini, T.; Vessecchi, R.; Pinto, E.; Collepicolo, P.; Lopes, N.P. Balance of xanthophylls molecular and protonated molecular ions in electrospray ionization. J. Mass Spectrom. 2005, 40, 963–968. [Google Scholar] [CrossRef]
  16. Bozin, B.; Mimica-Dukic, N.; Samojlik, I.; Jovin, E. Antimicrobial and Antioxidant Properties of Rosemary and Sage (Rosmarinus officinalis L. and Salvia officinalis L., Lamiaceae) Essential Oils. J. Agric. Food Chem. 2007, 55, 7879–7885. [Google Scholar] [CrossRef]
  17. Wang, W.; Li, N.; Luo, M.; Zu, Y.; Efferth, T. Antibacterial Activity and Anticancer Activity of Rosmarinus officinalis L. Essential Oil Compared to That of Its Main Components. Molecules 2012, 17, 2704–2713. [Google Scholar] [CrossRef] [Green Version]
  18. Moreno, S.; Scheyer, T.; Romano, C.S.; Vojnov, A.A. Antioxidant and Antimicrobial Activities of Rosemary Extracts Linked to Their Polyphenol Composition. Free. Radic. Res. 2006, 40, 223–231. [Google Scholar] [CrossRef] [PubMed]
  19. Waller, S.B.; Cleff, M.B.; Dalla Lana, D.F.; de Mattos, C.B.; Guterres, K.A.; Freitag, R.A.; Sallis, E.S.V.; Fuentefria, A.M.; de Mello, J.R.B.; de Faria, R.O.; et al. Can the Essential Oil of Rosemary (Rosmarinus officinalis Linn.) Protect Rats Infected with Itraconazole-Resistant Sporothrix Brasiliensis from Fungal Spread? J. Med. Mycol. 2021, 31, 101199. [Google Scholar] [CrossRef]
  20. Karpiński, T.M. Essential Oils of Lamiaceae Family Plants as Antifungals. Biomolecules 2020, 10, 103. [Google Scholar] [CrossRef] [Green Version]
  21. Lucarini, R.; Bernardes, W.A.; Ferreira, D.S.; Tozatti, M.G.; Furtado, R.; Bastos, J.K.; Pauletti, P.M.; Januário, A.H.; Silva, M.L.A.E.; Cunha, W.R. In Vivo Analgesic and Anti-Inflammatory Activities of Rosmarinus officinalis Aqueous Extracts, Rosmarinic Acid and Its Acetyl Ester Derivative. Pharm. Biol. 2013, 51, 1087–1090. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Ghasemzadeh Rahbardar, M.; Amin, B.; Mehri, S.; Mirnajafi-Zadeh, S.J.; Hosseinzadeh, H. Anti-Inflammatory Effects of Ethanolic Extract of Rosmarinus officinalis L. and Rosmarinic Acid in a Rat Model of Neuropathic Pain. Biomed. Pharmacother. 2017, 86, 441–449. [Google Scholar] [CrossRef] [PubMed]
  23. Hsieh, C.L.; Peng, C.H.; Chyau, C.C.; Lin, Y.C.; Wang, H.E.; Peng, R.Y. Low-Density Lipoprotein, Collagen, and Thrombin Models Reveal That Rosemarinus officinalis L. Exhibits Potent Antiglycative Effects. J. Agric. Food Chem. 2007, 55, 2884–2891. [Google Scholar] [CrossRef] [PubMed]
  24. Rodrigues, A.P.S.; Souza, B.S.F.E.; Barros, A.S.A.; de Oliveira Carvalho, H.; Duarte, J.L.; Boettger, L.E.M.; Barbosa, R.; Ferreira, A.M.; Ferreira, I.M.; Fernandes, C.P.; et al. The Effects of Rosmarinus officinalis L. Essential Oil and Its Nanoemulsion on Dyslipidemic Wistar Rats. J. Appl. Biomed. 2020, 18, 126–135. [Google Scholar] [CrossRef] [PubMed]
  25. López-Jiménez, A.; García-Caballero, M.; Medina, M.Á.; Quesada, A.R. Anti-Angiogenic Properties of Carnosol and Carnosic Acid, Two Major Dietary Compounds from Rosemary. Eur. J. Nutr. 2013, 52, 85–95. [Google Scholar] [CrossRef]
  26. Kayashima, T.; Matsubara, K. Antiangiogenic Effect of Carnosic Acid and Carnosol, Neuroprotective Compounds in Rosemary Leaves. Biosci. Biotechnol. Biochem. 2012, 76, 115–119. [Google Scholar] [CrossRef]
  27. Karthik, D.; Viswanathan, P.; Anuradha, C.V. Administration of Rosmarinic Acid Reduces Cardiopathology and Blood Pressure through Inhibition of P22phox NADPH Oxidase in Fructose-Fed Hypertensive Rats. J. Cardiovasc. Pharmacol. 2011, 58, 514–521. [Google Scholar] [CrossRef]
  28. Amaral, G.P.; de Carvalho, N.R.; Barcelos, R.P.; Dobrachinski, F.; de Lima Portella, R.; da Silva, M.H.; Lugokenski, T.H.; Dias, G.R.M.; da Luz, S.C.A.; Boligon, A.A.; et al. Protective Action of Ethanolic Extract of Rosmarinus officinalis L. in Gastric Ulcer Prevention Induced by Ethanol in Rats. Food Chem. Toxicol. 2013, 55, 48–55. [Google Scholar] [CrossRef]
  29. Samarghandian, S.; Borji, A.; Farkhondeh, T. Evaluation of Antidiabetic Activity of Carnosol (Phenolic Diterpene in Rosemary) in Streptozotocin-Induced Diabetic Rats. Cardiovasc. Haematol. Disord.-Drug Targets 2017, 17, 11–17. [Google Scholar] [CrossRef]
  30. Bakirel, T.; Bakirel, U.; Keleş, O.Ü.; Ülgen, S.G.; Yardibi, H. In Vivo Assessment of Antidiabetic and Antioxidant Activities of Rosemary (Rosmarinus officinalis) in Alloxan-Diabetic Rabbits. J. Ethnopharmacol. 2008, 116, 64–73. [Google Scholar] [CrossRef]
  31. Tai, J.; Cheung, S.; Wu, M.; Hasman, D. Antiproliferation Effect of Rosemary (Rosmarinus officinalis) on Human Ovarian Cancer Cells in Vitro. Phytomedicine 2012, 19, 436–443. [Google Scholar] [CrossRef] [PubMed]
  32. De Oliveira, J.R.; Camargo, S.E.A.; De Oliveira, L.D. Rosmarinus officinalis L. (Rosemary) as Therapeutic and Prophylactic Agent. J. Biomed. Sci. 2019, 26, 5. [Google Scholar] [CrossRef] [PubMed]
  33. Sharifi-Rad, J.; Ezzat, S.M.; El Bishbishy, M.H.; Mnayer, D.; Sharopov, F.; Kılıç, C.S.; Neagu, M.; Constantin, C.; Sharifi-Rad, M.; Atanassova, M.; et al. Rosmarinus Plants: Key Farm Concepts towards Food Applications. Phytother. Res. 2020, 34, 1474–1518. [Google Scholar] [CrossRef] [PubMed]
  34. Nieto, G.; Ros, G.; Castillo, J. Antioxidant and Antimicrobial Properties of Rosemary (Rosmarinus officinalis, L.): A Review. Medicines 2018, 5, 98. [Google Scholar] [CrossRef] [Green Version]
  35. Dorta, D.J.; Pigoso, A.A.; Mingatto, F.E.; Rodrigues, T.; Pestana, C.R.; Uyemura, S.A.; Santos, A.C.; Curti, C. Antioxidant Activity of Flavonoids in Isolated Mitochondria. Phytother. Res. 2008, 22, 1213–1218. [Google Scholar] [CrossRef] [PubMed]
  36. Okamura, N.; Haraguchi, H.; Hashimoto, K.; Yagi, A. Flavonoids in Rosmarinus officinalis Leaves. Phytochemistry 1994, 37, 1463–1466. [Google Scholar] [CrossRef]
  37. Forman, H.J.; Zhang, H. Targeting Oxidative Stress in Disease: Promise and Limitations of Antioxidant Therapy. Nat. Rev. Drug Discov. 2021, 20, 689–709. [Google Scholar] [CrossRef]
  38. El-Demerdash, F.M.; Abbady, E.A.; Baghdadi, H.H. Oxidative Stress Modulation by Rosmarinus officinalis in Creosote-Induced Hepatotoxicity. Environ. Toxicol. 2016, 31, 85–92. [Google Scholar] [CrossRef]
  39. Amin, A.; Hamza, A.A. Hepatoprotective Effects of Hibiscus, Rosmarinus and Salvia on Azathioprine-Induced Toxicity in Rats. Life Sci. 2005, 77, 266–278. [Google Scholar] [CrossRef]
  40. El-Demerdash, F.M.; El-Sayed, R.A.; Abdel-Daim, M.M. Hepatoprotective Potential of Rosmarinus officinalis Essential Oil against Hexavalent Chromium-Induced Hematotoxicity, Biochemical, Histological, and Immunohistochemical Changes in Male Rats. Environ. Sci. Pollut. Res. 2021, 28, 17445–17456. [Google Scholar] [CrossRef]
  41. Ramadan, K.S.; Khalil, O.A.; Danial, E.N.; Alnahdi, H.S.; Ayaz, N.O. Hypoglycemic and Hepatoprotective Activity of Rosmarinus officinalis Extract in Diabetic Rats. J. Physiol. Biochem. 2013, 69, 779–783. [Google Scholar] [CrossRef] [PubMed]
  42. El-Naggar, S.A.; Abdel-Farid, I.B.; Germoush, M.O.; Elgebaly, H.A.; Alm-Eldeen, A.A. Efficacy of Rosmarinus officinalis Leaves Extract against Cyclophosphamide-Induced Hepatotoxicity. Pharm. Biol. 2016, 54, 2007–2016. [Google Scholar] [CrossRef] [Green Version]
  43. Bunchorntavakul, C.; Reddy, K.R. Acetaminophen-Related Hepatotoxicity. Clin. Liver Dis. 2013, 17, 587–607. [Google Scholar] [CrossRef] [PubMed]
  44. Mazaleuskaya, L.L.; Sangkuhl, K.; Thorn, C.F.; Fitzgerald, G.A.; Altman, R.B.; Klein, T.E. PharmGKB Summary: Pathways of Acetaminophen Metabolism at the Therapeutic versus Toxic Doses. Pharm. Genom. 2015, 25, 416. [Google Scholar] [CrossRef] [Green Version]
  45. Figueira, T.R.; Barros, M.H.; Camargo, A.A.; Castilho, R.F.; Ferreira, J.C.B.; Kowaltowski, A.J.; Sluse, F.E.; Souza-Pinto, N.C.; Vercesi, A.E. Mitochondria as a Source of Reactive Oxygen and Nitrogen Species: From Molecular Mechanisms to Human Health. Antioxid. Redox Signal. 2013, 18, 2029–2074. [Google Scholar] [CrossRef]
  46. Sies, H. Oxidative Stress: Oxidants and Antioxidants. Exp. Physiol. 1997, 82, 291–295. [Google Scholar] [CrossRef]
  47. Cruz, T.S.; Faria, P.A.; Santana, D.P.; Ferreira, J.C.; Oliveira, V.; Nascimento, O.R.; Cerchiaro, G.; Curti, C.; Nantes, I.L.; Rodrigues, T. On the Mechanisms of Phenothiazine-Induced Mitochondrial Permeability Transition: Thiol Oxidation, Strict Ca2+ Dependence, and Cyt c Release. Biochem. Pharmacol. 2010, 80, 1284–1295. [Google Scholar] [CrossRef] [PubMed]
  48. Ainsworth, E.A.; Gillespie, K.M. Estimation of total phenolic content and other oxidation substrates in plant tissues using Folin-Ciocalteu reagent. Nat. Protoc. 2007, 2, 875–877. [Google Scholar] [CrossRef] [PubMed]
  49. Guimarães, N.S.S.; Mello, J.C.; Paiva, J.S.; Bueno, P.C.P.; Berretta, A.A.; Torquato, R.J.; Nantes, I.L.; Rodrigues, T. Baccharis dracunculifolia, the Main Source of Green Propolis, Exhibits Potent Antioxidant Activity and Prevents Oxidative Mitochondrial Damage. Food Chem. Toxicol. 2012, 50, 1091–1097. [Google Scholar] [CrossRef] [Green Version]
  50. Cowart, R.E.; Singleton, F.L.; Hind, J.S. A Comparison of Bathophenanthrolinedisulfonic Acid and Ferrozine as Chelators of Iron(II) in Reduction Reactions. Anal. Biochem. 1993, 211, 151–155. [Google Scholar] [CrossRef]
  51. Lebel, C.P.; Ischiropoulos, H.; Bondy, S.C. Evaluation of the probe 2′,7′-dichlorofluorescin as an indicator of reactive oxygen species formation and oxidative stress. Chem. Res. Toxicol. 1992, 5, 227–231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Mello, J.C.; Guimarães, N.S.S.; Gonzalez, M.V.D.; Paiva, J.S.; Prieto, T.; Nascimento, O.R.; Tiago Rodrigues, T. Hydroxyl scavenging activity accounts for differential antioxidant protection of Plantago major against oxidative toxicity in isolated rat liver mitochondria. J. Pharm. Pharmacol. 2012, 64, 1177–1187. [Google Scholar] [CrossRef] [PubMed]
  53. Mello, J.C.; Gonzalez, M.V.D.; Moraes, V.W.R.; Prieto, T.; Nascimento, O.R.; Rodrigues, T. Protective Effect of Plantago major Extract against t-BOOH-Induced Mitochondrial Oxidative Damage and Cytotoxicity. Molecules 2015, 20, 17747–17759. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Borges, M.B.D.; Dos Santos, C.G.; Yokomizo, C.H.; Sood, R.; Vitovic, P.; Kinnunen, P.K.J.; Rodrigues, T.; Nantes, I.L. Characterization of Hydrophobic Interaction and Antioxidant Properties of the Phenothiazine Nucleus in Mitochondrial and Model Membranes. Free. Radic. Res. 2010, 44, 1054–1063. [Google Scholar] [CrossRef] [PubMed]
  55. Hissin, P.J.; Hilf, R. A Fluorometric Method for Determination of Oxidized and Reduced Glutathione in Tissues. Anal. Biochem. 1976, 74, 214–226. [Google Scholar] [CrossRef]
  56. Cálgaro-Helena, A.F.; Devienne, K.F.; Rodrigues, T.; Dorta, D.J.; Raddi, M.S.G.; Vilegas, W.; Uyemura, S.A.; Santos, A.C.; Curti, C. Effects of Isocoumarins Isolated from Paepalanthus Bromelioides on Mitochondria: Uncoupling, and Induction/Inhibition of Mitochondrial Permeability Transition. Chem. Biol. Interact. 2006, 161, 155–164. [Google Scholar] [CrossRef]
  57. Jocelyn, P.C. Spectrophotometric Assay of Thiols. Methods Enzymol. 1987, 143, 44–67. [Google Scholar] [CrossRef]
  58. Mello, J.C.; Moraes, V.W.R.; Watashi, C.M.; Silva, D.C.; Cavalcanti, L.P.; Franco, M.K.K.D.; Yokaichiya, F.; Araujo, D.R.; Rodrigues, T. Enhancement of chlorpromazine antitumor activity by Pluronics F127/L81 nanostructured system against human multidrug resistant leukemia. Pharmacol. Res. 2016, 111, 102–112. [Google Scholar] [CrossRef]
  59. Pilon, A.C.; Del Grande, M.; Silvério, M.R.S.; Silva, R.R.; Albernaz, L.C.; Vieira, P.C.; Lopes, J.L.C.; Espindola, L.S.; Lopes, N.P. Combination of GC-MS Molecular Networking and Larvicidal Effect against Aedes aegypti for the Discovery of Bioactive Substances in Commercial Essential Oils. Molecules 2022, 27, 1588. [Google Scholar] [CrossRef]
  60. Liu, Y.; Guo, M. Studies on Transition Metal-Quercetin Complexes Using Electrospray Ionization Tandem Mass Spectrometry. Molecules 2015, 20, 8583–8594. [Google Scholar] [CrossRef] [Green Version]
  61. Hyung, S.C.; Jun, W.K.; Cha, Y.N.; Kim, C. A Quantitative Nitroblue Tetrazolium Assay for Determining Intracellular Superoxide Anion Production in Phagocytic Cells. J. Immunoass. Immunochem. 2006, 27, 31–44. [Google Scholar] [CrossRef]
  62. Pessoto, F.S.; Faria, P.A.; Cunha, R.L.O.R.; Comasseto, J.V.; Rodrigues, T.; Nantes, I.L. Organotellurane-Promoted Mitochondrial Permeability Transition Concomitant with Membrane Lipid Protection against Oxidation. Chem. Res. Toxicol. 2007, 20, 1453–1461. [Google Scholar] [CrossRef] [PubMed]
  63. Simunkova, M.; Barbierikova, Z.; Jomova, K.; Hudecova, L.; Lauro, P.; Alwasel, S.H.; Alhazza, I.; Rhodes, C.J.; Valko, M. Antioxidant vs. Prooxidant Properties of the Flavonoid, Kaempferol, in the Presence of Cu(II) Ions: A ROS-Scavenging Activity, Fenton Reaction and DNA Damage Study. Int. J. Mol. Sci. 2021, 22, 1619. [Google Scholar] [CrossRef] [PubMed]
  64. Jeffery, E.H.; Haschek, W.M. Protection by dimethylsulfoxide against acetaminophen-induced hepatic, but not respiratory toxicity in the mouse. Toxicol. Appl. Pharmacol. 1988, 93, 452–461. [Google Scholar] [CrossRef]
  65. Göksel Sener, G.; Sehirli, O.; Cetinel, S.; Yeğen, B.G.; Gedik, N.; Ayanoğlu-Dülger, G. Protective effects of MESNA (2-mercaptoethane sulphonate) against acetaminophen-induced hepatorenal oxidative damage in mice. J. Appl. Toxicol. 2005, 25, 20–29. [Google Scholar] [CrossRef]
  66. Lopes, N.P.; Stark, C.B.W.; Gates, P.J.; Staunton, J. Fragmentation studies on monensin A by sequential electrospray mass spectrometry. Analyst 2002, 127, 503–506. [Google Scholar] [CrossRef]
  67. Pilon, A.C.; Gu, H.; Raftery, D.; Bolzani, V.D.S.; Lopes, N.P.; Castro-Gamboa, I.; Carnevale Neto, F. Mass Spectral Similarity Networking and Gas-Phase Fragmentation Reactions in the Structural Analysis of Flavonoid Glycoconjugates. Anal. Chem. 2019, 91, 10413–10423. [Google Scholar] [CrossRef]
  68. Mena, P.; Cirlini, M.; Tassotti, M.; Herrlinger, K.A.; Dall’Asta, C.; Del Rio, D. Phytochemical Profiling of Flavonoids, Phenolic Acids, Terpenoids, and Volatile Fraction of a Rosemary (Rosmarinus officinalis L.) Extract. Molecules 2016, 21, 1576. [Google Scholar] [CrossRef]
  69. Sharma, Y.; Velamuri, R.; Fagan, J.; Schaefer, J. Full-Spectrum Analysis of Bioactive Compounds in Rosemary (Rosmarinus officinalis L.) as Influenced by Different Extraction Methods. Molecules 2020, 25, 4599. [Google Scholar] [CrossRef]
  70. Kontogianni, V.G.; Tomic, G.; Nikolic, I.; Nerantzaki, A.; Sayyad, A.; Stosic-Grujicic, N.; Stojanovic, S.; Gerothanassis, I.P.; Tzakos, A.G. Phytochemical profile of Rosmarinus officinalis and Salvia officinalis extracts and correlation to their antioxidant and anti-proliferative activity. Food Chem. 2013, 136, 120–129. [Google Scholar] [CrossRef]
  71. Wang, M.; Carver, J.J.; Phelan, V.V.; Sanchez, L.M.; Garg, N.; Peng, Y.; Nguyen, D.D.; Watrous, J.; Kapono, C.A.; Luzzatto-Knaan, T.; et al. Sharing and community curation of mass spectrometry data with Global Natural Products Social Molecular Networking. Nat. Biotechnol. 2016, 34, 828–837. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Green, D.R.; Reed, J.C. Mitochondria and Apoptosis. Science 1998, 281, 118. [Google Scholar] [CrossRef] [PubMed]
  73. Aruoma, O.I.; Halliwell, B.; Aeschbach, R.; Löligers, J. Antioxidant and Pro-Oxidant Properties of Active Rosemary Constituents: Carnosol and Carnosic Acid. Xenobiotica 1992, 22, 257–268. [Google Scholar] [CrossRef]
  74. Pérez-Fons, L.; Garzón, M.T.; Micol, V. Relationship between the Antioxidant Capacity and Effect of Rosemary (Rosmarinus officinalis L.) Polyphenols on Membrane Phospholipid Order. J. Agric. Food Chem. 2010, 58, 161–171. [Google Scholar] [CrossRef]
  75. Harlina, P.W.; Ma, M.; Shahzad, R.; Khalifa, I. Effect of Rosemary Extract on Lipid Oxidation, Fatty Acid Composition, Antioxidant Capacity, and Volatile Compounds of Salted Duck Eggs. Food Sci. Anim. Resour. 2022, 42, 689–711. [Google Scholar] [CrossRef]
  76. Nazem, F.; Farhangi, N.; Neshat-Gharamaleki, M. Beneficial Effects of Endurance Exercise with Rosmarinus officinalis Labiatae Leaves Extract on Blood Antioxidant Enzyme Activities and Lipid Peroxidation in Streptozotocin-Induced Diabetic Rats. Can. J. Diabetes 2015, 39, 229–234. [Google Scholar] [CrossRef] [PubMed]
  77. Karimi, G.; Hassanzadeh, M.; Mehri, S. Protective Effect of Rosmarinus officinalis L. Essential Oil against Free Radical-Induced Erythrocyte Lysis. Iran. J. Pharm. Sci. 2005, 1, 231–236. [Google Scholar]
  78. Kaliora, A.C.; Andrikopoulos, N.K. Effect of Alkanna Albugam Root on LDL Oxidation. A Comparative Study with Species of the Lamiaceae Family. Phytother. Res. 2005, 19, 1077–1079. [Google Scholar] [CrossRef]
  79. Haraguchi, H.; Saito, T.; Okamura, N.; Yagi, A. Inhibition of Lipid Peroxidation and Superoxide Generation by Diterpenoids from Rosmarinus officinalis. Planta Med. 1995, 61, 333–336. [Google Scholar] [CrossRef]
  80. Sueishi, Y.; Sue, M.; Masamoto, H. Seasonal Variations of Oxygen Radical Scavenging Ability in Rosemary Leaf Extract. Food Chem. 2018, 245, 270–274. [Google Scholar] [CrossRef]
  81. Horvathova, E.; Navarova, J.; Galova, E.; Sevcovicova, A.; Chodakova, L.; Snahnicanova, Z.; Melusova, M.; Kozics, K.; Slamenova, D. Assessment of Antioxidative, Chelating, and DNA-Protective Effects of Selected Essential Oil Components (Eugenol, Carvacrol, Thymol, Borneol, Eucalyptol) of Plants and Intact Rosmarinus officinalis Oil. J. Agric. Food Chem. 2014, 62, 6632–6639. [Google Scholar] [CrossRef]
  82. Lin, C.; Zhang, X.; Su, Z.; Xiao, J.; Lv, M.; Cao, Y.; Chen, Y. Carnosol Improved Lifespan and Healthspan by Promoting Antioxidant Capacity in Caenorhabditis elegans. Oxidative Med. Cell. Longev. 2019, 2019, 59580431619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Moreira, E.A.; Pilon, A.C.; Andrade, L.E.; Lopes, N.P. New Perspectives on Chlorogenic Acid Accumulation in Harvested Leaf Tissue: Impact on Traditional Medicine Preparations. ACS Omega 2018, 3, 12. [Google Scholar] [CrossRef]
  84. Zegura, B.; Dobnik, D.; Niderl, M.H.; Filipi, M. Antioxidant and antigenotoxic effects of rosemary (Rosmarinus officinalis L.) extracts in Salmonella typhimurium TA98 and HepG2 cells. Environ. Toxicol. Pharmacol. 2011, 32, 296–305. [Google Scholar] [CrossRef] [PubMed]
  85. Visentin, A.; Rodríguez-Rojo, S.; Navarrete, A.; Maestri, D.; Cocero, M.J. Precipitation and encapsulation of rosemary antioxidants by supercritical antisolvent process. J. Food Eng. 2012, 109, 9–15. [Google Scholar] [CrossRef]
  86. Sasaki, K.; El Omri, A.; Kondo, S.; Han, J.; Isoda, H. Rosmarinus officinalis polyphenols produce anti-depressant like effect through monoaminergic and cholinergic functions modulation. Behav. Brain Res. 2013, 238, 86–94. [Google Scholar] [CrossRef]
  87. Soares, M.S.; da Silva, D.F.; Forim, M.R.; da Silva, M.F.; Fernandes, J.B.; Vieira, P.C.; Silva, D.B.; Lopes, N.P.; de Carvalho, S.A.; de Souza, A.A.; et al. Quantification and localization of hesperidin and rutin in Citrus sinensis grafted on C. limonia after Xylella fastidiosa infection by HPLC-UV and MALDI imaging mass spectrometry. Phytochemistry 2015, 115, 161–170. [Google Scholar] [CrossRef] [PubMed]
  88. Kluska, M.; Juszczak, M.; Żuchowski, J.; Stochmal, A.; Woźniak, K. Effect of Kaempferol and Its Glycoside Derivatives on Antioxidant Status of HL-60 Cells Treated with Etoposide. Molecules 2022, 27, 333. [Google Scholar] [CrossRef] [PubMed]
  89. Hassanen, N.H.M.; Fahmi, A.; Shams-Eldin, E.; Abdur-Rahman, M. Protective Effect of Rosemary (Rosmarinus officinalis) against Diethylnitrosamine-Induced Renal Injury in Rats. Biomarkers 2020, 25, 281–289. [Google Scholar] [CrossRef]
  90. El-Demerdash, F.M.; El-Sayed, R.A.; Abdel-Daim, M.M. Rosmarinus officinalis Essential Oil Modulates Renal Toxicity and Oxidative Stress Induced by Potassium Dichromate in Rats. J. Trace Elem. Med. Biol. 2021, 67, 126791. [Google Scholar] [CrossRef]
  91. Rašković, A.; Milanović, I.; Pavlović, N.; Ćebović, T.; Vukmirović, S.; Mikov, M. Antioxidant Activity of Rosemary (Rosmarinus officinalis L.) Essential Oil and Its Hepatoprotective Potential. BMC Complement. Med. Ther. 2014, 14, 225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Sotelo-Félix, J.I.; Martinez-Fong, D.; Muriel, P.; Santillán, R.L.; Castillo, D.; Yahuaca, P. Evaluation of the Effectiveness of Rosmarinus officinalis (Lamiaceae) in the Alleviation of Carbon Tetrachloride-Induced Acute Hepatotoxicity in the Rat. J. Ethnopharmacol. 2002, 81, 145–154. [Google Scholar] [CrossRef] [PubMed]
  93. Gutiérrez, R.; Alvarado, J.L.; Presno, M.; Pérez-Veyna, O.; Serrano, C.J.; Yahuaca, P. Oxidative Stress Modulation by Rosmarinus officinalis in CCl4-Induced Liver Cirrhosis. Phytother. Res. 2010, 24, 595–601. [Google Scholar] [CrossRef]
  94. Ielciu, I.; Sevastre, B.; Olah, N.K.; Turdean, A.; Chișe, E.; Marica, R.; Oniga, I.; Uifălean, A.; Sevastre-Berghian, A.C.; Niculae, M.; et al. Evaluation of Hepatoprotective Activity and Oxidative Stress Reduction of Rosmarinus officinalis L. Shoots Tincture in Rats with Experimentally Induced Hepatotoxicity. Molecules 2021, 26, 1737. [Google Scholar] [CrossRef] [PubMed]
  95. Joyeux, M.; Rolland, A.; Fleurentin, J.; Mortier, F.; Dorfman, P. Tert-Butyl Hydroperoxide-Induced Injury in Isolated Rat Hepatocytes: A Model for Studying Anti-Hepatotoxic Crude Drugs. Planta Med. 1990, 56, 171–174. [Google Scholar] [CrossRef]
  96. Siemionow, K.; Teul, J.; Drągowski, P.; Pałka, J.; Miltyk, W. New Potential Biomarkers of Acetaminophen-Induced Hepatotoxicity. Adv. Med. Sci. 2016, 61, 325–330. [Google Scholar] [CrossRef]
  97. Botros, M.; Sikaris, K.A. The De Ritis Ratio: The Test of Time. Clin. Biochem. Rev. 2013, 34, 117. [Google Scholar]
  98. Corcoran, G.B.; Wong, B.K. Role of Glutathione in Prevention of Acetaminophen-Induced Hepatotoxicity by N-Acetyl-L-Cysteine in Vivo: Studies with N-Acetyl-D-Cysteine in Mice. J. Pharmacol. Exp. Ther. 1986, 238, 54–61. [Google Scholar]
  99. Hjelle, J.J.; Brzeznicka, E.A.; Klaassen, C.D. Comparison of the Effects of Sodium Sulfate and N-Acetylcysteine on the Hepatotoxicity of Acetaminophen in Mice. J. Pharmacol. Exp. Ther. 1986, 236, 526–534. [Google Scholar]
  100. Perron, N.R.; Brumaghim, J.L. A Review of the Antioxidant Mechanisms of Polyphenol Compounds Related to Iron Binding. Cell Biochem. Biophys. 2009, 53, 75–100. [Google Scholar] [CrossRef]
Figure 1. Glycolic extract of Rosmarinus officinalis (Ro) protects isolated rat liver mitochondria from oxidative stress. Mitochondria (1.0 mg/mL) were energized by succinate (site II substrate) in the presence of rotenone. DCF fluorescence was recorded kinetically using 0.6 mM t-BOOH ((A), red line) or 50 mM Fe2+ ((B), black line) as inducers of oxidative stress. Ro concentrations are represented by cyan (0.005%), blue (0.025%), and magenta (0.05%) lines. The inhibitory effect of Ro on the lipid oxidation (TBARS) induced by Fe2+ (squares, orange line) or t-BOOH (circles, red line). Effects of different concentrations of Ro on the formation of lipid hydroperoxides (LOOH) induced by Fe2+ (D), GSH oxidation induced by t-BOI (E), and oxidation of protein thiol groups induced by t-BOOH (F) or Fe2+ (G). In (DG), negative control (absence of Fe2+ and Ro) is represented by white bars, positive control (Fe2+ or t-BOOH) by black bars, and the effects of Ro by yellow (0.005%), orange (0.025%), and red (0.05%) bars. In (CG), data are presented as mean ± SD of three independent experiments. Statistical differences are indicated by * (p < 0.05) and ** (p < 0.01).
Figure 1. Glycolic extract of Rosmarinus officinalis (Ro) protects isolated rat liver mitochondria from oxidative stress. Mitochondria (1.0 mg/mL) were energized by succinate (site II substrate) in the presence of rotenone. DCF fluorescence was recorded kinetically using 0.6 mM t-BOOH ((A), red line) or 50 mM Fe2+ ((B), black line) as inducers of oxidative stress. Ro concentrations are represented by cyan (0.005%), blue (0.025%), and magenta (0.05%) lines. The inhibitory effect of Ro on the lipid oxidation (TBARS) induced by Fe2+ (squares, orange line) or t-BOOH (circles, red line). Effects of different concentrations of Ro on the formation of lipid hydroperoxides (LOOH) induced by Fe2+ (D), GSH oxidation induced by t-BOI (E), and oxidation of protein thiol groups induced by t-BOOH (F) or Fe2+ (G). In (DG), negative control (absence of Fe2+ and Ro) is represented by white bars, positive control (Fe2+ or t-BOOH) by black bars, and the effects of Ro by yellow (0.005%), orange (0.025%), and red (0.05%) bars. In (CG), data are presented as mean ± SD of three independent experiments. Statistical differences are indicated by * (p < 0.05) and ** (p < 0.01).
Antioxidants 12 00628 g001
Figure 2. Fe2+ chelating and free radical scavenger activities of glycolic extract of Rosmarinus officinalis (Ro). (A) 0.005 or 0.01% Ro were incubated with 50 μM Fe(NH4)2(SO4)2 and the absorbance of bathophenanthroline-Fe2+ complex was determined at 535 nm. (B) DPPH reduction by different Ro concentrations and the absorbance were determined at 517 nm, using 10 μM quercetin (Q) as a reference. (C) Superoxide was detected by its reaction with nitroblue tetrazolium (NBT) at 540 nm, considered 100% in the absence of Ro; different indicated Ro concentrations decreased NBT reduction. (D) Inhibitory effect of Ro on the lipid oxidation (TBARS) of 1.0 mM PCPECL liposomes induced by Fe2+. Results expressed as percentage were calculated in relation to control (absence of Ro or Fe2+), considered to be 100%. Data are presented as mean ± SD of three independent experiments. Statistical differences are indicated by ** (p < 0.01) and *** (p < 0.001).
Figure 2. Fe2+ chelating and free radical scavenger activities of glycolic extract of Rosmarinus officinalis (Ro). (A) 0.005 or 0.01% Ro were incubated with 50 μM Fe(NH4)2(SO4)2 and the absorbance of bathophenanthroline-Fe2+ complex was determined at 535 nm. (B) DPPH reduction by different Ro concentrations and the absorbance were determined at 517 nm, using 10 μM quercetin (Q) as a reference. (C) Superoxide was detected by its reaction with nitroblue tetrazolium (NBT) at 540 nm, considered 100% in the absence of Ro; different indicated Ro concentrations decreased NBT reduction. (D) Inhibitory effect of Ro on the lipid oxidation (TBARS) of 1.0 mM PCPECL liposomes induced by Fe2+. Results expressed as percentage were calculated in relation to control (absence of Ro or Fe2+), considered to be 100%. Data are presented as mean ± SD of three independent experiments. Statistical differences are indicated by ** (p < 0.01) and *** (p < 0.001).
Antioxidants 12 00628 g002
Figure 3. Absence of cytotoxicity of glycolic extract of Rosmarinus officinalis (Ro). (A) Viability of BRL 3A cells incubated with Ro (0 to 0.05%) for 24 h was estimated with MTT (A) and trypan blue (B) assays. The effects of Ro on cell viability were calculated in relation to control (absence of Ro), considered 100%. (C) Representative images of the mitochondrial membrane potential estimated by fluorescence microscopy using DiOC6(3). Cells were loaded with dyes, and fluorescence emission was acquired before and after 0.05% Ro addition. The uncoupler CCCP (10 μM) was used as positive control (see Methods for details). Scale bars (20 μm) (D) Relative quantification of DiOC6(3) fluorescence emission (L5 channel) considering the replicates. In (A,B) data are expressed as percentages of viable cells (mean ± SD) calculated in relation to control (i.e., without Ro), considered as 100%. Statistically non-significant (n.s.), considering p < 0.05.
Figure 3. Absence of cytotoxicity of glycolic extract of Rosmarinus officinalis (Ro). (A) Viability of BRL 3A cells incubated with Ro (0 to 0.05%) for 24 h was estimated with MTT (A) and trypan blue (B) assays. The effects of Ro on cell viability were calculated in relation to control (absence of Ro), considered 100%. (C) Representative images of the mitochondrial membrane potential estimated by fluorescence microscopy using DiOC6(3). Cells were loaded with dyes, and fluorescence emission was acquired before and after 0.05% Ro addition. The uncoupler CCCP (10 μM) was used as positive control (see Methods for details). Scale bars (20 μm) (D) Relative quantification of DiOC6(3) fluorescence emission (L5 channel) considering the replicates. In (A,B) data are expressed as percentages of viable cells (mean ± SD) calculated in relation to control (i.e., without Ro), considered as 100%. Statistically non-significant (n.s.), considering p < 0.05.
Antioxidants 12 00628 g003
Figure 4. Rosmarinus officinalis (Ro) protects against acetaminophen-induced hepatotoxicity in vivo. Rats were divided into four groups (n = 7) and treated intraperitoneally, as described in Materials and Methods. Group C (white bars): control, treated with vehicle (DMSO); group Ro (orange bars): 0.5% Ro once a day for four days; group AAP (black bars): single dose of 900 mg/kg acetaminophen; group Ro/AAP (red bars): received Ro for four days, followed by single dose of AAP. (A) Alanine aminotransferase and (B) aspartate aminotransferase levels. (C) GSH, (D) TBARS, and (E) reduced thiol groups of proteins quantified as described by isolated mitochondria in Figure 1. Statistical differences are indicated by * (p < 0.05), ** (p < 0.01), and *** (p < 0.001).
Figure 4. Rosmarinus officinalis (Ro) protects against acetaminophen-induced hepatotoxicity in vivo. Rats were divided into four groups (n = 7) and treated intraperitoneally, as described in Materials and Methods. Group C (white bars): control, treated with vehicle (DMSO); group Ro (orange bars): 0.5% Ro once a day for four days; group AAP (black bars): single dose of 900 mg/kg acetaminophen; group Ro/AAP (red bars): received Ro for four days, followed by single dose of AAP. (A) Alanine aminotransferase and (B) aspartate aminotransferase levels. (C) GSH, (D) TBARS, and (E) reduced thiol groups of proteins quantified as described by isolated mitochondria in Figure 1. Statistical differences are indicated by * (p < 0.05), ** (p < 0.01), and *** (p < 0.001).
Antioxidants 12 00628 g004
Figure 5. Chemical structures of compounds annotated in the Ro extract. Numbers in each structure refer to those presented in Table 2.
Figure 5. Chemical structures of compounds annotated in the Ro extract. Numbers in each structure refer to those presented in Table 2.
Antioxidants 12 00628 g005
Figure 6. Total Ion Chromatogram (TIC) of Ro obtained by LC-ESI-MS/MS analysis (negative mode). a Annotations from LC-ESI-MS/MS analysis in positive mode.
Figure 6. Total Ion Chromatogram (TIC) of Ro obtained by LC-ESI-MS/MS analysis (negative mode). a Annotations from LC-ESI-MS/MS analysis in positive mode.
Antioxidants 12 00628 g006
Table 1. Total phenols and flavonoids concentration present in 0.05% Ro.
Table 1. Total phenols and flavonoids concentration present in 0.05% Ro.
Concentration (μM)
Total phenols12.4 ± 0.2
Flavonoids7.2 ± 0.1
Table 2. Metabolites of Ro annotated by HPLC-ESI-MS/MS.
Table 2. Metabolites of Ro annotated by HPLC-ESI-MS/MS.
IDRT (min)MS
m/z [M − H]
MS/MS
m/z
CompoundReference
14.9179135Caffeic acid[68]
2a6.5305225Gallocatechin[69]
38.2237179, 161Caffeoylglycolic acid-
4a9.4359161(100), 179, 197Rosmarinic acid[70]
510.8461285(100), 199, 151Luteolin-7-O-glucuronide[68]
611.7503399, 285(100), 255Luteolin-3′-O-(3-acetyl)-glucuronide[11]
712.0315300(100), 227, 199Methoxy-tetra-hydroxy-flavone-
8a12.8345301, 283, 267, 217Rosmanol isomer[70]
9a13.4345283, 267, 227Rosmanol isomer/epirosmanol[70]
1014.1313298, 283, 255, 227, 164Cirsimaritin[71] (CCMSLIB00004718157)
1115.6345283, 227Rosmanol isomer/epirosmanol[70]
12a16.9343299, 243, 216Rosmadial or Rosmanol quinone [68]
13a17.7329285(100), 201Carnosol[70]
1418.9315285, 201Rosmaridiphenol[68]
1520.3331287(100), 272, 244Carnosic acid[68]
1620.9345301(100), 286, 271Methyl carnosate[70]
RT (min)MS
m/z [M + H]+
MS/MS
m/z
CompoundReference
1710.3611547, 449, 287(100)Hesperidin[71] (CCMSLIB00000214340)
1810.8449287(100)Astragalin[71] (CCMSLIB00003136634)
a Majority compounds—TIC (negative mode).
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

Guimarães, N.S.S.; Ramos, V.S.; Prado-Souza, L.F.L.; Lopes, R.M.; Arini, G.S.; Feitosa, L.G.P.; Silva, R.R.; Nantes, I.L.; Damasceno, D.C.; Lopes, N.P.; et al. Rosemary (Rosmarinus officinalis L.) Glycolic Extract Protects Liver Mitochondria from Oxidative Damage and Prevents Acetaminophen-Induced Hepatotoxicity. Antioxidants 2023, 12, 628. https://0-doi-org.brum.beds.ac.uk/10.3390/antiox12030628

AMA Style

Guimarães NSS, Ramos VS, Prado-Souza LFL, Lopes RM, Arini GS, Feitosa LGP, Silva RR, Nantes IL, Damasceno DC, Lopes NP, et al. Rosemary (Rosmarinus officinalis L.) Glycolic Extract Protects Liver Mitochondria from Oxidative Damage and Prevents Acetaminophen-Induced Hepatotoxicity. Antioxidants. 2023; 12(3):628. https://0-doi-org.brum.beds.ac.uk/10.3390/antiox12030628

Chicago/Turabian Style

Guimarães, Natalia S. S., Vyctória S. Ramos, Laura F. L. Prado-Souza, Rayssa M. Lopes, Gabriel S. Arini, Luís G. P. Feitosa, Ricardo R. Silva, Iseli L. Nantes, Debora C. Damasceno, Norberto P. Lopes, and et al. 2023. "Rosemary (Rosmarinus officinalis L.) Glycolic Extract Protects Liver Mitochondria from Oxidative Damage and Prevents Acetaminophen-Induced Hepatotoxicity" Antioxidants 12, no. 3: 628. https://0-doi-org.brum.beds.ac.uk/10.3390/antiox12030628

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

Article Metrics

Back to TopTop