Next Article in Journal
Androgen Receptor in Hormone Receptor-Positive Breast Cancer
Next Article in Special Issue
Progress in Predicting Ames Test Outcomes from Chemical Structures: An In-Depth Re-Evaluation of Models from the 1st and 2nd Ames/QSAR International Challenge Projects
Previous Article in Journal
Increased Mortality Risk at Septic Condition in Inflammatory Skin Disorders and the Effect of High-Fat Diet Consumption
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

In Vivo and In Silico Studies of the Hepatoprotective Activity of Tert-Butylhydroquinone

by
Liseth Rubi Aldaba-Muruato
1,
Sandra Sánchez-Barbosa
1,
Víctor Hugo Rodríguez-Purata
2,
Georgina Cabrera-Cruz
1,
Estefany Rosales-Domínguez
1,
Daniela Martínez-Valentín
1,
Yoshio Aldo Alarcón-López
3,
Pablo Aguirre-Vidal
3,
Manuel Alejandro Hernández-Serda
3,
Luis Alfonso Cárdenas-Granados
3,
Víctor Hugo Vázquez-Valadez
3,
Enrique Angeles
3 and
José Roberto Macías-Pérez
1,*
1
Biomedical Science Laboratory, Clinical Chemistry, Faculty of Professional Studies Huasteca Zone, Autonomous University of San Luis Potosi, Ciudad Valles 79060, San Luis Potosi, Mexico
2
Pharmacobiological Sciences, Faculty of Chemical Sciences, Autonomous University of San Luis Potosi, San Luis Potosi 78210, Mexico
3
Laboratorio de Química Teórica y Medicinal, FESC, Universidad Nacional Autónoma de México, Avenida 1 de Mayo S/N, Santa María las Torre, Cuautitlán Izcalli 54750, Estado de México, Mexico
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(1), 475; https://doi.org/10.3390/ijms25010475
Submission received: 16 November 2023 / Revised: 14 December 2023 / Accepted: 23 December 2023 / Published: 29 December 2023

Abstract

:
Tert-butylhydroquinone (TBHQ) is a synthetic food antioxidant with biological activities, but little is known about its pharmacological benefits in liver disease. Therefore, this work aimed to evaluate TBHQ during acute liver damage induced by CCl4 (24 h) or BDL (48 h) in Wistar rats. It was found that pretreatment with TBHQ prevents 50% of mortality induced by a lethal dose of CCl4 (4 g/kg, i.p.), and 80% of BDL+TBHQ rats survived, while only 50% of the BDL group survived. Serum markers of liver damage and macroscopic and microscopic (H&E staining) observations suggest that TBHQ protects from both hepatocellular necrosis caused by the sublethal dose of CCl4 (1.6 g/kg, i.p.), as well as necrosis/ductal proliferation caused by BDL. Additionally, online databases identified 49 potential protein targets for TBHQ. Finally, a biological target candidate (Keap1) was evaluated in a proof-of-concept in silico molecular docking assay, resulting in an interaction energy of −5.5491 kcal/mol, which was higher than RA839 and lower than monoethyl fumarate (compounds known to bind to Keap1). These findings suggest that TBHQ increases the survival of animals subjected to CCl4 intoxication or BDL, presumably by reducing hepatocellular damage, probably due to the interaction of TBHQ with Keap1.

1. Introduction

Worldwide, there are approximately two million deaths caused by liver disease, of which half are from cirrhosis and the other half from viral hepatitis and hepatocarcinoma [1]. The liver is a multifunctional organ susceptible to damage due to several factors, such as the consumption of alcohol or drugs, obstruction of bile flow, exposure to toxins, or parasitic and viral infections [2,3,4]. Certainly, regardless of the causative agent of liver damage, hepatocyte cell death favors the progression of the disease, starting with inflammation that leads to fibrosis and finally to liver cancer [3,5]. However, despite medical advances, there are no effective treatments that can prevent or even reverse liver disease, with organ transplantation being the procedure of choice for irreversible liver damage. However, there is a growing number of patients waiting for a liver transplant that exceed the number of donors [6].
Liver pathogenesis manifests itself as inflammation and necrosis, cholestasis, fibrosis, cirrhosis, and hepatocarcinoma [1,7]. In the present investigation, the hepatoprotective capacity of Tert-butylhydroquinone (TBHQ) was evaluated during acute liver damage induced by extrahepatic cholestasis and carbon tetrachloride (CCl4) intoxication, and its possible mechanism of protection was hypothesized by bioinformatic and in silico analysis.
TBHQ (CH3)3CC6H3-1,4-(OH)2 is a crystalline powder, a member of the class of hydroquinones in which one of the hydrogens in the hydroquinone ring is replaced by a tert-butyl group (Figure 1). This compound is commonly used as a synthetic food antioxidant to prevent oxidative deterioration and the rancidity of oils and fats due to its potent activity to inhibit lipid peroxidation [8,9]. Recent studies attribute various biological activities, such as the preservation of testicular steroidogenesis and spermatogenesis, to improving ethanol-induced gastric ulcers and radiocontrast-induced nephropathy [10,11,12].
The bioactive effect of TBHQ is attributed to the induction of phase II drug-metabolizing enzymes through a dependent pathway of nuclear factor erythroid 2-related factor 2 (Nrf2), which is a member of the basic leucine zipper transcription factor (bZIP), regulated mainly by Kelch-like ECH-associated protein 1 (Keap1) [13]. Keap1 is an adapter substrate of the Cullin–RING E3 ubiquitin ligase complex, which suppresses Nrf2 in the cytoplasm by sequestration, ubiquitination, and proteasomal degradation [14]. Under the stimulus of reactive oxygen species (ROS) or electrophilic agents, the hyperreactive cysteine residues of Keap1 allow the inactivation of the E3 ubiquitin ligase, and Nrf2 is dissociated from Keap1. Nrf2 accumulates in the cytoplasm and travels to the nucleus to mediate cytoprotective gene expression through antioxidant-responsive elements (AREs). The Keap1 protein contains a domain called BTB (bric-a-brac) that mediates dimerization, another domain called the IVR (intermediate region) domain that is involved with the interaction with CUL3, and a joint domain called DC composed of the individual domains DGR (double glycine repeat) and CTR (carboxyl-terminal region). In this DC domain, the interaction with Nrf2 occurs. On the Nrf2 protein, there are DLG and ETGE motifs in the section called Neh2 (amino end) that interact with the DC domains of the Keap1 homodimer [15,16]. Therefore, another objective of the present study was to analyze the interaction between TBHQ and Keap1 in silico (Figure 2).

2. Results

2.1. Mortality and Survival

The CCl4 model allowed us to evaluate the protective capacity of TBHQ against lethal and sublethal toxicity induced by CCl4 (Table 1). After 24 h, a lethal dose of CCl4 (4 g/kg, i.p.) yielded 100% (6/6) mortality, while TBHQ pretreatment prevented 50% (3/6) of mortality induced by the lethal dose. However, all animals injected with a sublethal dose of CCl4 (1.6 g/kg, i.p.) survived. In addition, bile duct ligation (BDL group, 48 h) leads to 50% mortality and pretreatment with TBHQ shows a mortality of 20% (80% survival).

2.2. Body and Liver Weights

Statistical analysis of body weights, liver weights, and liver/body ratio was performed in both experimental models of liver damage (Table 2). Body weight was significantly reduced in the CCl4+TBHQ group in relation to the CCl4 and TBHQ groups, while liver weight increased in the CCl4 and CCl4+TBHQ groups compared to the Control group. Furthermore, liver/body weight increased significantly in the CCl4 and CCl4+TBHQ groups compared to the TBHQ and Control groups. However, the SHAM (simulated surgery), BDL, BDL+TBHQ, and TBHQ groups did not show statistically significant differences between them for any of the weights.

2.3. Macroscopic Findings of Livers

Livers were observed in situ (Figure 3), and healthy control groups (Control, TBHQ, SHAM, and SHAM+TBHQ) presented a uniform and smooth surface with soft consistency on palpation. Additionally, livers in the BDL group showed an altered appearance with a pale surface, while the CCl4 group presented an altered hepatic morphology with a necrotic appearance. The gross appearance of the livers from the CCl4+TBHQ and BDL+TBHQ groups showed a less damaged appearance compared to the damage groups (CCl4 or BDL, respectively).

2.4. Microscopic Evaluation of Liver Damage Induced by a Sublethal Dose of CCl4

The hepatoprotective activity of TBHQ was evaluated during the sublethal dose of CCl4 with hematoxylin–eosin (H&E) staining (Figure 4). The hepatic parenchyma of the Control and TBHQ groups showed a normal architecture characteristic of healthy livers, with normal organization of hepatocytes organized in cords, in addition to clearly identifying some central veins and hepatic sinusoids, as shown in the micrographs. On the other hand, micrographs of the CCl4 group showed severe damage with evident steatosis and inflammatory infiltrate with ballooned hepatocytes; however, in liver samples of the CCl4+TBHQ group, less steatosis and ballooning degeneration were observed, showing better architectural integrity than the CCl4 group, whose damage was greater.

2.5. Microscopic Evaluation of Liver Damage Induced by BDL

H&E staining showed that two days after surgery, the BDL group presented liver necrosis, with a loss of normal architecture of the liver cords and infiltration of inflammatory cells with marked proliferation of the bile ducts. However, treatment with TBHQ prevented acute liver damage induced by BDL, since the BDL+TBHQ group showed little infiltration of inflammatory cells and mild proliferation of the bile ducts, as well as less hepatic necrosis and preservation of cell integrity, which can be compared to that of the healthy groups (Figure 5).

2.6. Serum Biochemical Markers of Liver Damage

Hepatic functionality was evaluated using the serum enzymatic activities of alanine aminotransferase (ALT), gamma-glutamyl transpeptidase (GGT), and alkaline phosphatase (ALP) (Figure 6). Intoxicated animals with a sublethal dose of CCl4 (Figure 6a–c) or with biliary obstruction (Figure 6d–f) showed significant increases in ALT, ALP, and GGT, respectively. Compared to the CCl4 group, TBHQ treatment (CCl4+TBHQ) partially but significantly prevented ALT (Figure 6a) and completely prevented increases in GGT (Figure 6b) and ALP (Figure 6c), similarly, to the Control group. Similarly, BDL and TBHQ partially prevent the increase in enzymatic activity of ALT, GGT, and ALP. The healthy groups (Control, TBHQ, SHAM, and SHAM+TBHQ) did not show significant differences between them in the enzymatic markers evaluated.

2.7. Identification of Potential TBHQ Protein Targets in the Liver

The possible mechanism of action of TBHQ was hypothesized using online platforms to search for its potential protein targets and data curated from the public Human Protein Atlas database. Thus, 49 potential targets for the THBQ protein were chosen (Table 3) if readily indicated as liver, expressed in liver, enriched in hepatocytes, liver stellate cells, or Kupffer cells (liver-specific macrophages), or were subunits of nuclear factor-kappa B (NF-κB), which is known to be involved in liver damage [17].

2.8. Molecular Docking of TBHQ and Keap1

The molecular systems found in the PDB files mentioned above showed that the amino acids in Keap1 interact with the residues Arg415, Ala556, Arg483, Arg483, Tyr334, Ser363, and Ser602 of (3S)-1-[4-[(2,3,5,6-tetramethylphenyl) sulfonylamino] -1-naphthyl] pyrrolidine-3-carboxylic acid, named as RA839 (Figure 7a). Examination showed that the amino acids in Keap1 that interact with monoethyl fumarate (MEF) are present in two regions: the first consisted near the residues Gln530, Tyr525, and Tyr572 (Figure 7b) and near the residues Asp422, Gly423 and Gly371, Gly372, Val369, and Val370 (Figure 7c). The information obtained from the Keap1 complex of Keap1 and the N-terminal region of Nrf2 showed that the Keap1 residues involved in the interaction are Arg415, Arg483, Ser508, Tyr525, Gln530, Ser555, Ala556, Tyr572, and Phe577 (Figure 7d).
Based on the previously described examination, we identified the common residues in the interaction present in each complex between the molecule and Nrf2. In the case of RA839, the Keap1 amino acids Arg 415, Arg483, and Ala556 are also involved in the interaction between Keap1 and Nrf2. For MEF, the Keap1 amino acids Gln530, Tyr525, and Tyr572 are also involved in the interaction of Keap1 with Nrf2. Using SiteFinder, a potential binding site was found near the residues shown in Table 4 and Figure 8, highlighting the common residues involved in the interaction of the PDB files.
During the docking experiments, the three cocrystallized ligands were re-evaluated to assess their energetic binding affinity. The interaction energy between the ligand obtained from PDB 7K2M, corresponding to the Nef2 domain of Nrf2, was observed to be the highest of all. The interaction of the MEF (from PDB 7C60) and RA839 (from PDB 5CGJ) molecules has lower interaction energies. When comparing the resulting interaction energy for TBHQ, it was higher than that of MEF but lower than that of RA839 (Table 5).
Here, we examined the modeled interaction between Keap1 and TBHQ. At the modeled site, the ligand is deeply incorporated into the channel that Keap1 forms. This explains the greater binding affinity of the ligand‒receptor complex. If we look inside the 2D interactions, we can see that the contour (dotted line in Figure 9) covers most of the molecular surface of TBHQ and that the ligand exposure (marked with faded purple circles) is small. The strongest interaction is due to the hydrogen bonding of one hydroxyl group with Gly-367. The rest of the interactions responsible for stabilizing the complex are due to electrostatic interactions, protein surface complementarity, and contact preferences. The nearest residues are Gly367, Val604, Gly605, Gly464, Gly 558, and Val418, among others.
Although the site of interaction of this simulated complex is far from the X-ray structures that we found and described above, the size of the molecule could be an explanation for the position of the ligand in the protein. Considering that a channel is formed in the structure of Keap1 and due to the size of TBHQ, after replication of the simulation, TBHQ was always allocated deep into the channel, maintaining good binding energies (Figure 10).

3. Discussion

Approximately 2 million people die from liver disease each year due to the poor effectiveness of therapeutic treatments, leading to increased morbidity and mortality rates around the world [1]. Therefore, it is necessary to search for new compounds that can mitigate, prevent, or reverse liver damage. In the present work, we evaluated TBHQ as a possible hepatoprotective agent due to its anti-inflammatory, anti-apoptotic, and antioxidant properties that have been previously described [18,19,20]. Similarly, various in vitro and in vivo studies have demonstrated that TBHQ exhibits chemopreventive effects, although this compound has also been described as carcinogenic, though mainly when used at high concentrations [13]. In the CCl4 model used in the present work, two doses of TBHQ were used: 40 mg/kg, i.p., and 16.7 mg/kg, i.p., initially, TBHQ was administered for two days at 40 mg/kg, i.p., daily [21,22]; this dose in in vivo models increases the long-term stability of curcumin [21] and leads to induction of UGT1A1, through the Nrf2–Keap1 pathway [22]. However, on the second day, immediately after administration, animals became lethargic for 5 to 10 min and recovered (at this dose and the route of administration in experimental animals, we were unable to find these side effects reported in the literature). Taking into account these side effects, the dose was reduced to 16.7 mg/kg, i.p., every 8 h [19], and no adverse effects were observed in any of the animals. This dose of TBHQ reduced ischemia/reperfusion injury in diabetic rats [19]. Therefore, further toxicological tests should be performed. However, it was conclusive that the mortality of the animals was reduced when they were pretreated with TBHQ (Table 1).
Rats in the BDL group had 50% mortality in 48 h, while the TBHQ+BDL group had 20% mortality with a survival rate of 80%. Furthermore, CCl4 at 4 g/kg, i.p., is lethal to 100% of rats intoxicated but pretreatment with TBHQ reduces this mortality by 50%. Common bile duct ligation induces liver damage by accumulating bile acids [23]. The obstruction of bile flow induces severe liver damage, so the mortality rate in experimental animals is very high [24]. Furthermore, cholestasis increases mortality and morbidity in patients undergoing major liver surgery because it is associated with increased sepsis and ischemia/reperfusion injury in the liver, accompanied by endothelial damage, inflammation, increased reactive oxygen species and proinflammatory cytokines, and activation of coagulation and fibrinolysis [25]. Therefore, our results suggest that TBHQ may be a suitable candidate to try to reduce complications related to cholestasis. Furthermore, chronic cholestatic liver diseases induce progressive hepatobiliary damage, with subsequent complications such as fibrosis and cirrhosis, ultimately leading to cancer [5]. Clinically, obstruction of the bile duct generates jaundice, choluria, and hepatomegaly, and, biochemically, it increases plasma liver enzyme markers [26,27]. All animals with obstruction of the bile duct (BDL and BDL+TBHQ groups) presented jaundice (as well as choluria results not shown); micrographs of the BDL group showed necrosis and duct proliferation, although hepatomegaly was not evident (Figure 5, Table 2). The BDL+TBHQ group showed a more preserved macroscopic and microscopic morphology (Figure 3 and Figure 6) than the BDL group. These results were consistent with serum biochemical analyses (Figure 6); animals partially but significantly prevented enzymatic increases in the hepatocellular necrosis marker (ALT) and the cholestasis marker (ALP or GGT). These findings suggest for the first time that TBHQ protects against liver injury induced by obstructive cholestasis.
On the other hand, CCl4 is a chlorinated hydrocarbon that was anciently used as a degreaser in household cleaning, industrial factories, dry cleaners, and textile laundries. It was also used in fire extinguishers and as a precursor to refrigerants and propellants, but due to its high oxidative toxicity, it has fallen into disuse; however, some industries still use it [28]. Currently, it is used in scientific research with experimental animals, emulating acute or chronic hepatocellular damage in humans, allowing the evaluation of new strategies to prevent or reverse hepatocellular damage [23]. This toxic solvent is metabolized by cytochrome p450, mainly by CYP2E1, producing free radicals such as CCl3* and CCl2* that affect lipid metabolism by inhibiting its transport out of hepatocytes and, in turn, increasing lipid synthesis, leading to steatosis [28]. In the present study, we observed steatohepatitis in the groups of rats intoxicated with a sublethal dose of CCl4, with extensive areas of ballooning hepatocellular degeneration (Figure 4), previously characterized by enlarged hepatocytes with intermediate filaments embedded in clear cytoplasm in combination with lobular inflammation, necrosis, and steatosis [29]; these morphologic alterations were reduced in the CCl4+TBHQ group. In macroscopic analyses, it can be seen that the liver weight/body weight ratio (Table 2) of the CCl4 and CCl4+TBHQ groups increased considerably, presenting statistically significant differences with respect to the Control group. The higher this value is, the larger the livers are compared to the weight of the rat, indicating the possible presence of hepatomegaly, a very common alteration in livers affected by CCl4 metabolism [30]. The inflammation process is a defense mechanism with the aim of limiting and eliminating causes of cell damage, as well as facilitating the tissue repair process. Acute and chronic liver damage is caused by acute inflammation, which is self-limited, contrary to prolonged chronic inflammation common in many diseases, such as cirrhosis [31]. Some studies in animal models of liver damage have described that TBHQ is a hepatoprotective agent, such as in the study carried out in male C57bl/6 mice, which showed that TBHQ mitigates acute liver damage induced by CCl4 at a dose of 0.1 mL/kg, i.p. (10 mL/kg body weight volume CCl4/volume olive oil = 1:99); another work showed that TBHQ exerts anti-inflammatory activity in liver injury ischemia and reperfusion in male Sprague‒Dawley rats [32,33]. In addition, some in vitro studies suggest that TBHQ at high doses (100 and 500 µM) is cytotoxic in human and murine hepatoma cell lines, respectively [13]. Furthermore, a recent review described the beneficial and toxic effects of TBHQ, suggesting that more research on public health and the mechanism of action in different organs and cells should be carefully explored [34]. Therefore, the present work made a comparison of two different types of liver damage, demonstrating for the first time the anticholestatic effect of TBHQ.
Because TBHQ presents a wide variety of biological effects [10,11,12], it is possible that it has multiple molecular targets in addition to the already known targets; therefore, online platforms were consulted to predict its possible molecular targets (Table 3). Fifty-six proteins were identified, of which two were transcription factors: Nrf2 (Protein ID: NFE2L2) and p65 (Protein ID: RELA). TBHQ has been associated with the activation of the Nrf2 pathway in HepG2 cells [32] and protects against ischemia/reperfusion-induced liver injury through the activation of the Keap1/Nrf2/ARE signaling pathway in rats [33]. Keap1 represses the Nrf2/ARE pathway in two ways: in the cytoplasm, Keap1 recruits Nrf2 into the Cul3/containing E3 ubiquitin ligase complex of Cul3/containing E3, causing its proteasomal degradation; however, Keap1 is able to translocate to the nucleus to dissociate Nrf2 from ARE [35]. Furthermore, the activation of the Nrf2/ARE signaling pathway induced by TBHQ is inhibited by the NF-κB subunit p65 through its interaction with Keap1, which induces the nuclear export of Nrf2 [36,37]. In the present work, the interaction between TBHQ and Keap1 was hypothesized.
Furthermore, in Table 3, other target proteins were identified, such as the nuclear factor-kappa B p105 subunit (Protein ID: NFKB1), which is a suppressor of inflammation [38]; the nuclear receptor ROR gamma liver (Protein ID: RORC), considered a potential therapeutic target in liver fibrosis [39]; xanthine dehydrogenase (ID: XDH), which catalyzes the oxidation of NADH-generating tissue injuries mediated by reactive oxygen species [40]; and the bile acid receptor (Protein ID: NR1H4), whose low expression is related to biliary atresia characterized by fibrous obstruction in childhood liver diseases [41]. These, among other molecules, make up 96.4% of the identified protein targets, for which the effects of TBHQ are unknown. The meticulous analyses in this table open up the possibility of further studies involving these proteins in liver disease.

4. Materials and Methods

4.1. Animals

A total of 36 male Wistar rats weighing between 200 and 250 g were used in the CCl4 model and 40 male Wistar rats weighing 300 to 350 g were used in the bile duct ligation model (BDL). All rats were kept with ad libitum water and a standard diet (LabDiet 5008) [42] and 12 h of light and 12 h of darkness at 24 °C. Experimental procedures in rats were approved by the Research Ethics Committee of the Huasteca Zone Faculty of Professional Studies of the Autonomous University of San Luis Potosí, México, and were carried out based on international terms and guidelines, as well as technical specifications for the production, care, and use of laboratory animals dictated by the official Mexican standard NOM-062-ZOO-1999 [43].

4.2. Preparation of TBHQ

To be administered orally, 100 mg of TBHQ (Sigma‒Aldrich, Saint Louis, MO, USA) was dissolved in 100 μL of dimethylsulfoxide (DMSO, Sigma‒Aldrich, Saint Louis, MO, USA) and then diluted with buffered sterile phosphate saline (PBS, pH 7.4), with constant stirring in the dark to a final concentration of 5 mg/mL TBHQ in 1% DMSO and administered immediately after preparation.

4.3. Experimental Protocol of the CCl4 Model

4.3.1. Administration of TBHQ to Rats Intoxicated with CCl4

Pretreatments with TBHQ (40 mg/kg, i.p., every 8 h) [21,22] started three days before CCl4 intoxication [19].

4.3.2. Mortality Assessment during the Lethal Dose of CCl4

Twelve rats were divided into two groups (Figure 11). CCl4 group (n = 6): lethal dose of CCl4 (4 g/kg, i.p.) [44] and TBHQ+CCl4 group (n = 6): TBHQ+CCl4 (4 g/kg, i.p.). Mortality was recorded 24 h after CCl4 administration.

4.3.3. Hepatoprotective Activity of TBHQ during the Sublethal Dose of CCl4

Twenty-four rats were divided into four groups (Figure 12). Control group (n = 6), CCl4 group (n = 6): sublethal dose of 1.6 g/kg, i.p., of CCl4 [45,46], TBHQ+CCl4 group (n = 6): TBHQ+CCl4 (1.6 g/kg, i.p.), and TBHQ group. Mineral oil (CCl4 vehicle) was administered i.p. to the Control and TBHQ groups. Survival was recorded after 24 h of CCl4 or mineral oil administration.

4.4. Experimental Protocol of the BDL Model

The rats were divided into four groups (Figure 13). BDL group (n = 10): rats were previously administered 1% DMSO (1 mL, i.p., every administration, every 8 h) for a total of four doses before surgery, which consisted of performing a double ligation in the common bile duct, one close to the duodenum and another proximal to the liver; finally, the bile duct was cut off [26,47] and administration of 1% DMSO continued every 8 h until sacrifice. BDL+TBHQ group (n = 10): rats were pretreated 24 h before BDL with three oral administrations of TBHQ (16.7 mg/kg, i.p., every 8 h) and the fourth administration was carried out 30 min before the surgical procedure. After surgery, the administrations continued every 8 h until sacrifice. SHAM group (n = 10): simulated surgery rats were treated with 1% DMSO. SHAM+TBHQ group (n = 10): animals with SHAM surgery and TBHQ administration. Mortality in rats from the acute liver cholestasis model was recorded 48 h after undergoing SHAM or BDL surgical procedures. Finally, all animals were sacrificed 48 h after SHAM or BDL surgeries.

4.5. Sacrifice of Experimental Animals

The rats were weighed and immediately anesthetized with an i.p. mixture of xylazine hydrochloride (Sigma‒Aldrich) at 10 mg/kg and ketamine hydrochloride (Sigma‒Aldrich) at 80 mg/kg and immediately sacrificed by cardiac puncture. The blood collected was centrifuged (1000× g, 10 min, 4 °C) to obtain serum. Furthermore, the liver was dissected, washed in a cold saline solution, and weighed. Small liver fragments were then obtained and immersed in 4% p-formaldehyde, pH 7.0.

4.6. H&E Staining Procedure

Liver tissues fixed for 48 h with 4% p-formaldehyde pH 7.0 were progressively dehydrated with organic solvents as follows (1 h each): 70% ethanol, 80% ethanol, 96% ethanol, absolute ethanol, ethanol-xylene 1:1, xylene (twice), and hot paraffin (twice). Next, tissues were embedded in paraffin, and 4 µm histological sections were prepared on an Ecoshel model 202A microtome and fixed on slides treated with 2% 3-aminopropyltriethoxysilane in acetone [48]. Next, they were subjected to deparaffinization at 60 °C for 24 h. H&E staining was carried out in the following order: xylol 10 min, xylol 1 min, absolute ethanol-xylol (1:1) 1 min, absolute ethanol 1 min, ethanol 96% 1 min, ethanol 80% 1 min, hematoxylin 5 min, wash with tap water 6 min, acid alcohol 0.5% 1 s, distilled water 1 min, ammonia solution 0.05% 5 min, distilled water 1 min, distilled water 1 min, eosin 1 min, 80% ethanol 1 min, 96% ethanol 1 min, absolute ethanol 1 min, absolute ethanol-xylene (1:1) 1 min, and twice with xylene (1 min each).

4.7. Photographic Images of H&E Staining

H&E stains were visualized with a Zeiss Axioscope 40/40 FL microscope and analyzed with ImageJ version 1.53e software.

4.8. Evaluation of Serum Biochemical Markers of Liver Damage

The enzyme activities of ALT [49], GGT [50], and ALP [51] were evaluated in serum samples. Briefly, plasma ALT enzyme activity was evaluated in duplicate for each test: 250 μL of substrate solution (0.2 M D/L of alanine with 2 M α-ketoglutaric acid) and 50 μL of serum were mixed and incubated at 37 °C for 60 min. Subsequently, 250 μL of the chromogenic reagent (1 mM 2,4-dinitrophenylhydrazine) was added and the sample was further incubated for 15 min at the same temperature. Finally, 1.5 mL of 0.4 N NaOH was added and measured on a spectrophotometer at a wavelength of 515 nm. The enzyme activity of ALP was determined in each sample in duplicate by adding 250 μL of 0.1 M glycine buffer, 1 mM MgCl2 with a pH of 10.5, and 250 μL of p-nitrophenylphosphate substrate, mixing and incubating at 37 °C for 5 min. After that time, 50 μL of serum was added to incubate again at 37 °C for 30 min. Finally, NaOH 0.02 N was added and absorbances were measured in a spectrophotometer with a wavelength of 410 nm. The GGT enzymatic activities were carried out in duplicate in each sample with 400 μL of 0.2 M Tris-HCl reagent, 100 μL of MgCl2, 100 μL of 0.04 M glycyl-glycine, and 100 μL of 10 mM gamma-glutamyl-p-nitroanilide. Once the solution was prepared, it was incubated for 10 min at 37 °C, after which 200 μL of serum to evaluate was added, and it was incubated again for 30 min at the same temperature of 37 °C. After incubation, 2 mL of 1.5 M acetic acid was added to stop the reaction, and the absorbance was measured in a spectrophotometer at a wavelength of 410 nm. For the three markers of liver damage, blanks were included, and the respective standard curves were performed as suggested by the authors.

4.9. Statistical Analysis

Statistical analysis was performed using GraphPad Prism 8.00 software. The results of the biochemical studies were expressed as the mean values ± SE from each experimental group, and comparative analysis was carried out using variance analysis followed by Tukey’s test. Statistical significance was considered at p < 0.05.

4.10. Identification of Potential Targets for TBHQ Protein in the Liver

Eight online platforms, where protein targets for chemical compounds can be searched or predicted, were consulted to analyze the types of proteins with which TBHQ could interact. The platforms used were ChEMBL [52,53], PharmMapper [54,55,56], Pharos [57], PPB [58], SEA [59], Super-PRED [60], SwissTargetPrediction [61], and TargetNet [62], and they were consulted between 5 and 14 July 2022. On these platforms, the TBHQ molecule was submitted either by drawing it or in mol2 or SMILES formats, with both generated on the ChemInfo website using OpenBabel software [63,64], where the molecule was drawn to obtain them. From each set of data obtained, the protein targets with the highest probability of being the said targets were chosen, according to the platform algorithm and if indicated in the results (PharmMapper, Super-PRED, SwissTargetPrediction, and TargetNet), or all those obtained by the other platforms were further used. Of the protein targets chosen, their protein code was searched in the GeneCards database [65,66] if it was not listed in the results of each platform. With this code, the Human Protein Atlas, a large compendium that integrates results from omics technologies to map human protein expression in cells, tissues, and organs, was consulted [67].

4.11. Molecular Docking of TBHQ with Keap1

4.11.1. Keap1/Nrf2 Protein

The Nrf2 is a transcription factor with seven domains: Neh 1, Neh 2, Neh 3, Neh 4, Neh 5, Neh 6, and Neh 7 (Table 6 and Figure 14).

4.11.2. Molecular Docking

We analyzed the interaction of TBHQ by looking for molecules that inhibit the interaction between Nrf2 and Keap1, since there are crystallized structures of Keap1 with small molecules in the interaction domain between them that may function as controls for this putative interaction. First, PDB 5CGJ, a Keap1 crystal structure of Keap1, bound to RA839, a small molecule that binds noncovalently to the Keap1–Kelch domain and affects its interaction with Nrf2 (Kd = 6 μM) [68]. This structure has an X-ray diffraction resolution of 3.36 Å, an R-value of 0.226, and an R-value of 0.137. A preliminary analysis identified the amino acids in Keap1 that interact with RA839. The second PDB used was 7C60, which contains the Keap1 crystal in a complex with MEF, which is the metabolite of the Nrf2-activating drug dimethyl fumarate used for the treatment of multiple sclerosis [69]. This structure has an X-ray resolution of 1.95 Å, an R-value of 0.288, and an R-value of 0.214. A preliminary analysis was performed to identify amino acid residues involved in the interaction between Keap1 and MEF. Finally, the interaction between Keap1 and Nrf2 was screened using PDB 7K2M, which has a resolution of 2.02 Å, an R-value of 0.261, and an R-value of 0.237 [70]. Preliminarily, we analyzed which Keap1 residues are involved in this interaction.
Molecular docking analysis was prepared and performed with the best-resolved Keap1 protein from PDB 7C60 using MOE 2022.02 (Chemical Computing Group, Molecular Operating Environment (MOE), 2022.02 Chemical Computing Group ULC, 1010 Sherbrooke St. West, Suite #910, Montreal, QC, Canada, H3A 2R7, 2022). The protein was parameterized using the AMBER 14: EHT forcefield, water molecules were removed, and the structure was minimized. Subsequently, the cocrystallized ligands were removed. By using SiteFinder from MOE, a site was found where some of the already identified residues (shown in Figure 8) and the tested small molecules (RA839, MEF, and TBHQ) could be hosted, and a set of dummy atoms was placed near the site. With this information, that site was used as a receptor for molecular docking. The receptor was programmed to be rigid, and the search algorithm was based on the geometry of Triangle Matcher with London dG as the scoring function to obtain 100 poses. The protocol was followed by a pose refinement using the GBVI/WSA dG scoring function to filter 10 of the most energetically favorable conformations of protein‒ligand complexes [71,72,73,74,75,76,77,78,79].

5. Conclusions

Our results suggest for the first time that the administration of TBHQ increases survival in animals with extrahepatic cholestasis or by lethal toxicity of CCl4 and propose new protein targets that can be evaluated as possible protection mechanisms for TBHQ against liver disease. In our proof-of-concept model, we found that TBHQ has a tendency to introduce deeper into the protein structure compared to RA839, MEF, and the Nrf2–Kelch domain, which explains its binding energy being greater than MEF but still lower than RA839; nevertheless, the interaction and complementarity of TBHQ in Keap1 could have a major influence on inhibition of the Keap1–Nrf2–Keap1 forming trimer Keap1–Nrf2–Keap1, which could cause an increase in Nrf2 concentration into the cytoplasm to be translocated to the nucleus, then activating the expression of antioxidant proteins responsible for the hepatoprotective effect. Even when TBHQ could not interact directly with the residues involved in the formation of the Keap1–Nrf2 complex, based on the idea that even a small change in the geometry of the binding site could cause a large change in protein conformation, the deeper introduction of our ligand into Keap1 could cause an impediment to Nrf2 binding and then increase its concentration, causing the abovementioned effect.

Author Contributions

Conceptualization, J.R.M.-P. and L.R.A.-M.; methodology G.C.-C., D.M.-V., V.H.R.-P., E.R.-D., Y.A.A.-L., P.A.-V., M.A.H.-S., L.A.C.-G., V.H.V.-V., J.R.M.-P. and L.R.A.-M.; software, S.S.-B. and L.R.A.-M.; validation, J.R.M.-P., E.A., S.S.-B. and L.R.A.-M.; formal analysis, J.R.M.-P., E.A. and L.R.A.-M.; investigation, J.R.M.-P., E.A. and L.R.A.-M.; resources, J.R.M.-P. and E.A.; data curation, E.A., J.R.M.-P. and L.R.A.-M.; writing—original draft preparation, L.R.A.-M.; writing—review and editing, E.A., J.R.M.-P., Y.A.A.-L., P.A.-V., M.A.H.-S., L.A.C.-G., V.H.V.-V. and L.R.A.-M.; visualization, J.R.M.-P. and L.R.A.-M.; supervision, E.A., J.R.M.-P. and L.R.A.-M.; project administration, J.R.M.-P.; funding acquisition, J.R.M.-P. and E.A. All authors have read and agreed to the published version of the manuscript.

Funding

The authors wish to thank CONACYT (No. 320331 “Ciencia Básica y/o de Frontera Modalidad: Paradigmas y Controversias de la Ciencia 2022”) for their financial support. Furthermore, this project was partially supported by the UNAM PAPIIT IN206023, PIAPI2005, Grupo de Trabajo XVIII GTMQ de la SRE de México, and LANCAD-UNAM-DGTIC-034.

Institutional Review Board Statement

All experimental protocols using rats were approved by the Research Ethics Committee, Faculty of Professional Studies Huasteca Zone, Autonomous University of San Luis Potosí, and were conducted according to institutional guidelines. All animals received care according to international guidelines and in adherence to the technical specifications established in the official Mexican standard NOM-062-ZOO-1999.

Data Availability Statement

The data presented in this study are available on request from the corresponding author J.R.M.-P.

Acknowledgments

The authors thank Moises Hernández and Draucin Jiménez from FESC-UNAM for their technical assistance.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Asrani, S.K.; Devarbhavi, H.; Eaton, J.; Kamath, P.S. Burden of liver diseases in the world. J. Hepatol. 2019, 70, 151–171. [Google Scholar] [CrossRef] [PubMed]
  2. Wu, H.; Chen, C.; Ziani, S.; Nelson, L.J.; Ávila, M.A.; Nevzorova, Y.A.; Cubero, F.J. Fibrotic Events in the Progression of Cholestatic Liver Disease. Cells 2021, 10, 1107. [Google Scholar] [CrossRef] [PubMed]
  3. Shojaie, L.; Iorga, A.; Dara, L. Cell Death in Liver Diseases: A Review. Int. J. Mol. Sci. 2020, 21, 9682. [Google Scholar] [CrossRef] [PubMed]
  4. Ben-Moshe, S.; Itzkovitz, S. Spatial heterogeneity in the mammalian liver. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 395–410. [Google Scholar] [CrossRef] [PubMed]
  5. Palmer, M.; Regev, A.; Lindor, K.; Avigan, M.I.; Dimick-Santos, L.; Treem, W.; Marcinak, J.F.; Lewis, J.H.; Anania, F.A.; Seekins, D.; et al. Consensus guidelines: Best practices for detection, assessment and management of suspected acute drug-induced liver injury occurring during clinical trials in adults with chronic cholestatic liver disease. Aliment. Pharmacol. Ther. 2020, 51, 90–109. [Google Scholar] [CrossRef] [PubMed]
  6. Meirelles Júnior, R.F.; Salvalaggio, P.; Rezende, M.B.; Evangelista, A.S.; Guardia, B.D.; Matielo, C.E.; Neves, D.B.; Pandullo, F.L.; Felga, G.E.; Alves, J.A.; et al. Liver transplantation: History, outcomes and perspectives. Einstein 2015, 13, 149–152. [Google Scholar] [CrossRef] [PubMed]
  7. Ezhilarasan, D. Oxidative stress is bane in chronic liver diseases: Clinical and experimental perspective. Arab. J. Gastroenterol. 2018, 19, 56–64. [Google Scholar] [CrossRef]
  8. PubChem. Bethesda (MD): National Library of Medicine (US), National Center for Biotechnology Information; 2004. PubChem Compound Summary for CID 16043, Tert-Butylhydroquinone. Available online: https://pubchem.ncbi.nlm.nih.gov/compound/16043 (accessed on 6 October 2023).
  9. Farhoosh, R. Initiation and propagation kinetics of inhibited lipid peroxidation. Sci. Rep. 2021, 11, 6864. [Google Scholar] [CrossRef]
  10. Nna, V.U.; Ujah, G.A.; Suleiman, J.B.; Mohamed, M.; Nwokocha, C.; Akpan, T.J.; Ekuma, H.C.; Fubara, V.V.; Kekung-Asu, C.B.; Osim, E.E. Tert-butylhydroquinone preserve testicular steroidogenesis and spermatogenesis in cisplatin-intoxicated rats by targeting oxidative stress, inflammation and apoptosis. Toxicology 2020, 441, 152528. [Google Scholar] [CrossRef]
  11. Rahman, Z.; Dwivedi, D.K.; Jena, G.B. Ethanol-induced gastric ulcer in rats and intervention of tert-butylhydroquinone: Involvement of Nrf2/HO-1 signalling pathway. Hum. Exp. Toxicol. 2020, 39, 547–562. [Google Scholar] [CrossRef]
  12. Zhou, Q.; Wang, X.; Shao, X.; Wang, H.; Liu, X.; Ke, X.; Xiong, C.; Wei, L.; Zou, H. tert-Butylhydroquinone Treatment Alleviates Contrast-Induced Nephropathy in Rats by Activating the Nrf2/Sirt3/SOD2 Signaling Pathway. Oxid. Med. Cell. Longev. 2019, 2019, 4657651. [Google Scholar] [CrossRef] [PubMed]
  13. Gharavi, N.; Haggarty, S.; El-Kadi, A.O. Chemoprotective and carcinogenic effects of tert-butylhydroquinone and its metabolites. Curr. Drug Metab. 2007, 8, 1–7. [Google Scholar] [CrossRef] [PubMed]
  14. Silva-Islas, C.A.; Maldonado, P.D. Canonical and non-canonical mechanisms of Nrf2 activation. Pharmacol. Res. 2018, 134, 92–99. [Google Scholar] [CrossRef] [PubMed]
  15. Zhou, J.; Zheng, Q.; Chen, Z. The Nrf2 Pathway in Liver Diseases. Front. Cell Dev. Biol. 2022, 10, 826204. [Google Scholar] [CrossRef] [PubMed]
  16. Kensler, T.W.; Wakabayashi, N.; Biswal, S. Cell survival responses to environmental stresses via the Keap1-Nrf2-ARE pathway. Annu. Rev. Pharmacol. Toxicol. 2007, 47, 89–116. [Google Scholar] [CrossRef]
  17. Nowak, A.J.; Relja, B. The Impact of Acute or Chronic Alcohol Intake on the NF-κB Signaling Pathway in Alcohol-Related Liver Disease. Int. J. Mol. Sci. 2020, 21, 9407. [Google Scholar] [CrossRef]
  18. Bai, J.; Yu, X.J.; Liu, K.L.; Wang, F.F.; Li, H.B.; Shi, X.L.; Zhang, Y.; Huo, C.J.; Li, X.; Gao, H.L.; et al. Tert-butylhydroquinone attenuates oxidative stress and inflammation in hypothalamic paraventricular nucleus in high salt-induced hypertension. Toxicol. Lett. 2017, 281, 1–9. [Google Scholar] [CrossRef]
  19. Gong, D.J.; Wang, L.; Yang, Y.Y.; Zhang, J.J.; Liu, X.H. Diabetes aggravates renal ischemia and reperfusion injury in rats by exacerbating oxidative stress, inflammation, and apoptosis. Ren. Fail. 2019, 41, 750–761. [Google Scholar] [CrossRef]
  20. Veskemaa, L.; Graw, J.A.; Pickerodt, P.A.; Taher, M.; Boemke, W.; González-López, A.; Francis, R.C.E. Tert-butylhydroquinone augments Nrf2-dependent resilience against oxidative stress and improves survival of ventilator-induced lung injury in mice. Am. J. Physiol. Lung Cell. Mol. Physiol. 2021, 320, L17–L28. [Google Scholar] [CrossRef]
  21. Nimiya, Y.; Wang, W.; Du, Z.; Sukamtoh, E.; Zhu, J.; Decker, E.; Zhang, G. Redox modulation of curcumin stability: Redox active antioxidants increase chemical stability of curcumin. Mol. Nutr. Food Res. 2016, 60, 487–494. [Google Scholar] [CrossRef]
  22. Yueh, M.F.; Tukey, R.H. Nrf2-Keap1 signaling pathway regulates human UGT1A1 expression in vitro and in transgenic UGT1 mice. J. Biol. Chem. 2007, 282, 8749–8758. [Google Scholar] [CrossRef] [PubMed]
  23. Muriel, P. Some Experimental Models of Liver Damage. In Hepatotoxicity; Sahu, S.C., Ed.; John Wiles & Sons, Ltd.: Hoboken, NJ, USA, 2008; pp. 119–137. [Google Scholar] [CrossRef]
  24. Tag, C.G.; Sauer-Lehnen, S.; Weiskirchen, S.; Borkham-Kamphorst, E.; Tolba, R.H.; Tacke, F.; Weiskirchen, R. Bile duct ligation in mice: Induction of inflammatory liver injury and fibrosis by obstructive cholestasis. J. Vis. Exp. 2015, 96, 52438. [Google Scholar] [CrossRef]
  25. Kloek, J.J.; Levi, M.; Heger, M.; van der Loos, C.M.; Gouma, D.J.; van Gulik, T.M. Cholestasis enhances liver ischemia/reperfusion-induced coagulation activation in rats. Hepatol. Res. 2010, 40, 204–215. [Google Scholar] [CrossRef] [PubMed]
  26. Van Campenhout, S.; Van Vlierberghe, H.; Devisscher, L. Common Bile Duct Ligation as Model for Secondary Biliary Cirrhosis. Methods Mol. Biol. 2019, 1981, 237–247. [Google Scholar] [CrossRef] [PubMed]
  27. Johnstone, J.M.; Lee, E.G. A quantitative assessment of the structural changes the rat’s liver following obstruction of the common bile duct. Br. J. Exp. Pathol. 1976, 57, 85–94. [Google Scholar] [PubMed]
  28. Al Amin, A.S.M.; Menezes, R.G. Carbon Tetrachloride Toxicity. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2023. Available online: https://www.ncbi.nlm.nih.gov/books/NBK562180/ (accessed on 1 March 2023).
  29. Lackner, C.; Gogg-Kamerer, M.; Zatloukal, K.; Stumptner, C.; Brunt, E.M.; Denk, H. Ballooned hepatocytes in steatohepatitis: The value of keratin immunohistochemistry for diagnosis. J. Hepatol. 2008, 48, 821–828. [Google Scholar] [CrossRef] [PubMed]
  30. Platt, D.S.; Cockrill, B.L. Liver enlargement and hepatoxicity: An investigation into the effects of several agents on rat liver enzyme activities. Biochem. Pharmacol. 1967, 16, 2257–2270. [Google Scholar] [CrossRef]
  31. Ahmed, S.M.; Luo, L.; Namani, A.; Wang, X.J.; Tang, X. Nrf2 signaling pathway: Pivotal roles in inflammation. Biochim. Biophys. 2017, 1863, 585–597. [Google Scholar] [CrossRef]
  32. Li, R.; Zhang, P.; Li, C.; Yang, W.; Yin, Y.; Tao, K. Tert-butylhydroquinone mitigates Carbon Tetrachloride induced Hepatic Injury in mice. Int. J. Med. Sci. 2020, 17, 2095–2103. [Google Scholar] [CrossRef]
  33. Zeng, X.P.; Li, X.J.; Zhang, Q.Y.; Liu, Q.W.; Li, L.; Xiong, Y.; He, C.X.; Wang, Y.F.; Ye, Q.F. Tert-Butylhydroquinone Protects Liver Against Ischemia/Reperfusion Injury in Rats Through Nrf2-Activating Anti-Oxidative Activity. Transplant. Proc. 2017, 49, 366–372. [Google Scholar] [CrossRef]
  34. Khezerlou, A.; Akhlaghi, A.P.; Alizadeh, A.M.; Dehghan, P.; Maleki, P. Alarming impact of the excessive use of tert-butylhydroquinone in food products: A narrative review. Toxicol. Rep. 2022, 9, 1066–1075. [Google Scholar] [CrossRef] [PubMed]
  35. Sun, Z.; Zhang, S.; Chan, J.Y.; Zhang, D.D. Keap1 controls postinduction repression of the Nrf2-mediated antioxidant response by escorting nuclear export of Nrf2. Mol. Cell. Biol. 2007, 27, 6334–6349. [Google Scholar] [CrossRef] [PubMed]
  36. Yu, M.; Li, H.; Liu, Q.; Liu, F.; Tang, L.; Li, C.; Yuan, Y.; Zhan, Y.; Xu, W.; Li, W.; et al. Nuclear factor p65 interacts with Keap1 to repress the Nrf2-ARE pathway. Cell. Signal. 2011, 23, 883–892. [Google Scholar] [CrossRef] [PubMed]
  37. Hu, B.; Wei, H.; Song, Y.; Chen, M.; Fan, Z.; Qiu, R.; Zhu, W.; Xu, W.; Wang, F. NF-κB and Keap1 Interaction Represses Nrf2-Mediated Antioxidant Response in Rabbit Hemorrhagic Disease Virus Infection. J. Virol. 2020, 94, e00016-20. [Google Scholar] [CrossRef] [PubMed]
  38. Cartwright, T.; Perkins, N.D.; Wilson, C.L. NFKB1: A suppressor of inflammation, ageing and cancer. FEBS J. 2016, 283, 1812–1822. [Google Scholar] [CrossRef]
  39. Kim, S.M.; Choi, J.E.; Hur, W.; Kim, J.H.; Hong, S.W.; Lee, E.B.; Lee, J.H.; Li, T.Z.; Sung, P.S.; Yoon, S.K. RAR-Related Orphan Receptor Gamma (ROR-γ) Mediates Epithelial-Mesenchymal Transition Of Hepatocytes During Hepatic Fibrosis. J. Cell. Biochem. 2017, 118, 2026–2036. [Google Scholar] [CrossRef]
  40. Maia, L.; Vala, A.; Mira, L. NADH oxidase activity of rat liver xanthine dehydrogenase and xanthine oxidase-contribution for damage mechanisms. Free Radic. Res. 2005, 39, 979–986. [Google Scholar] [CrossRef]
  41. Ma, Y.; Lu, L.; Tan, K.; Li, Z.; Guo, T.; Wu, Y.; Wu, W.; Zheng, L.; Fan, F.; Mo, J.; et al. Reduced peroxisome proliferator-activated receptor-α and bile acid nuclear receptor NR1H4/FXR may affect the hepatic immune microenvironment of biliary atresia. Front. Immunol. 2022, 13, 875593. [Google Scholar] [CrossRef]
  42. Gomez, C.B.; de la Cruz, S.H.; Medina-Terol, G.J.; Beltran-Ornelas, J.H.; Sánchez-López, A.; Silva-Velasco, D.L.; Centurión, D. Chronic administration of NaHS and L-Cysteine restores cardiovascular changes induced by high-fat diet in rats. Eur J Pharmacol. 2019, 863, 172707. [Google Scholar] [CrossRef]
  43. Norma Oficial Mexicana NOM-062-ZOO-1999: Especificaciones Técnicas para la Producción, Cuidado y Uso de los Animales de Laboratorio. 1999. Available online: https://www.fmvz.unam.mx/fmvz/principal/archivos/062ZOO.PDF (accessed on 27 February 2023).
  44. Tezuka, M.; Momiyama, K.; Edano, T.; Okada, S. Protective effect of chromium(III) on acute lethal toxicity of carbon tetrachloride in rats and mice. J. Inorg. Biochem. 1991, 42, 1–8. [Google Scholar] [CrossRef]
  45. Bakdemir, M.; Çetin, E. Hepatoprotective effects of ethyl pyruvate against carbon tetrachloride-induced oxidative stress, biochemical and histological alterations in rats. Arch. Physiol. Biochem. 2021, 127, 359–366. [Google Scholar] [CrossRef] [PubMed]
  46. Ohta, Y.; Kongo, M.; Sasaki, E.; Nishida, K.; Ishiguro, I. Therapeutic effect of melatonin on carbon tetrachloride-induced acute liver injury in rats. J. Pineal Res. 2000, 28, 119–126. [Google Scholar] [CrossRef] [PubMed]
  47. Georgiev, P.; Jochum, W.; Heinrich, S.; Jang, J.H.; Nocito, A.; Dahm, F.; Clavien, P.A. Characterization of time-related changes after experimental bile duct ligation. Br. J. Surg. 2008, 95, 646–656. [Google Scholar] [CrossRef] [PubMed]
  48. Aragón, P.; Noguera, P.; Bañuls, M.J.; Puchades, R.; Maquieira, Á.; González-Martínez, M.Á. Modulating receptor-ligand binding in biorecognition by setting surface wettability. Anal. Bioanal. Chem. 2018, 410, 5723–5730. [Google Scholar] [CrossRef] [PubMed]
  49. Reitman, S.; Frankel, S. A colorimetric method for the determination of serum glutamic oxalacetic and glutamic pyruvic transaminases. Am. J. Clin. Pathol. 1957, 28, 56–63. [Google Scholar] [CrossRef] [PubMed]
  50. Glossmann, H.; Neville, D.M. gamma-Glutamyltransferase in kidney brush border membranes. FEBS Lett. 1972, 19, 340–344. [Google Scholar] [CrossRef] [PubMed]
  51. Bergmeyer, H.U.; Grassl, M.; Walter, H.E. Methods of Enzymatic Analysis, 3rd ed.; Bergmeyer, H.U.M., Ed.; Verlag Chemie: Deerfield Beach, FL, USA, 1983; Volume 2, pp. 269–270. [Google Scholar]
  52. Davies, M.; Nowotka, M.; Papadatos, G.; Dedman, N.; Gaulton, A.; Atkinson, F.; Bellis, L.; Overington, J.P. ChEMBL web services: Streamlining access to drug discovery data and utilities. Nucleic Acids Res. 2015, 43, W612–W620. [Google Scholar] [CrossRef]
  53. Mendez, D.; Gaulton, A.; Bento, A.P.; Chambers, J.; De Veij, M.; Félix, E.; Magariños, M.P.; Mosquera, J.F.; Mutowo, P.; Nowotka, M.; et al. ChEMBL: Towards direct deposition of bioassay data. Nucleic Acids Res. 2019, 47, D930–D940. [Google Scholar] [CrossRef]
  54. Liu, X.; Ouyang, S.; Yu, B.; Liu, Y.; Huang, K.; Gong, J.; Zheng, S.; Li, Z.; Li, H.; Jiang, H. PharmMapper server: A web server for potential drug target identification using pharmacophore mapping approach. Nucleic Acids Res. 2010, 38, W609–W614. [Google Scholar] [CrossRef]
  55. Wang, X.; Pan, C.; Gong, J.; Liu, X.; Li, H. Enhancing the Enrichment of Pharmacophore-Based Target Prediction for the Polypharmacological Profiles of Drugs. J. Chem. Inf. Model. 2016, 56, 1175–1183. [Google Scholar] [CrossRef]
  56. Wang, X.; Shen, Y.; Wang, S.; Li, S.; Zhang, W.; Liu, X.; Lai, L.; Pei, J.; Li, H. PharmMapper 2017 update: A web server for potential drug target identification with a comprehensive target pharmacophore database. Nucleic Acids Res. 2017, 45, W356–W360. [Google Scholar] [CrossRef]
  57. Sheils, T.K.; Mathias, S.L.; Kelleher, K.J.; Siramshetty, V.B.; Nguyen, D.T.; Bologa, C.G.; Jensen, L.J.; Vidović, D.; Koleti, A.; Schürer, S.C.; et al. TCRD and Pharos 2021: Mining the human proteome for disease biology. Nucleic Acids Res. 2021, 49, D1334–D1346. [Google Scholar] [CrossRef] [PubMed]
  58. Reymond, J.L. PPB Polypharmacology Browser for Target Prediction in ChEMBL Browser. PPB. 13 July 2022. Available online: http://gdbtools.unibe.ch:8080/PPB/contact.html (accessed on 4 February 2023).
  59. Keiser, M.J.; Roth, B.L.; Armbruster, B.N.; Ernsberger, P.; Irwin, J.J.; Shoichet, B.K. Relating protein pharmacology by ligand chemistry. Nat. Biotechnol. 2007, 25, 197–206. [Google Scholar] [CrossRef] [PubMed]
  60. Nickel, J.; Gohlke, B.O.; Erehman, J.; Banerjee, P.; Rong, W.W.; Goede, A.; Dunkel, M.; Preissner, R. SuperPred: Update on drug classification and target prediction. Nucleic Acids Res. 2014, 42, W26–W31. [Google Scholar] [CrossRef] [PubMed]
  61. Daina, A.; Michielin, O.; Zoete, V. SwissTargetPrediction: Updated data and new features for efficient prediction of protein targets of small molecules. Nucleic Acids Res. 2019, 47, W357–W364. [Google Scholar] [CrossRef] [PubMed]
  62. Yao, Z.J.; Dong, J.; Che, Y.J.; Zhu, M.F.; Wen, M.; Wang, N.N.; Wang, S.; Lu, A.P.; Cao, D.S. TargetNet: A web service for predicting potential drug-target interaction profiling via multi-target SAR models. J. Comput. Aided. Mol. Des. 2016, 30, 413–424. [Google Scholar] [CrossRef] [PubMed]
  63. Cheminfo.org. OPENBABEL–Chemical File Format Converter. Cheminfo.org. 5 July 2022. Available online: http://www.cheminfo.org/Chemistry/Cheminformatics/FormatConverter/index.html# (accessed on 18 May 2023).
  64. O’Boyle, N.M.; Banck, M.; James, C.A.; Morley, C.; Vandermeersch, T.; Hutchison, G.R. Open Babel: An open chemical toolbox. J. Cheminform. 2011, 3, 33. [Google Scholar] [CrossRef]
  65. Lancet Lab at the Weizmann Institute of Science. GeneCards–The Human Gene Database. GeneCardsSuite. 14 July 2022. Available online: https://www.genecards.org/ (accessed on 18 May 2023).
  66. Safran, M.; Rosen, N.; Twik, M.; BarShir, R.; Stein, T.I.; Dahary, D.; Fishilevich, S.; Lancet, D. The GeneCards Suite. In Practical Guide to Life Science Databases, 1st ed.; Abugessaisa, I., Kasukawa, T., Eds.; Springer: Singapore, 2021; pp. 27–56. [Google Scholar]
  67. The Human Protein Atlas. Available online: https://www.proteinatlas.org/ (accessed on 10 June 2023).
  68. Winkel, A.F.; Engel, C.K.; Margerie, D.; Kannt, A.; Szillat, H.; Glombik, H.; Kallus, C.; Ruf, S.; Güssregen, S.; Riedel, J.; et al. Characterization of RA839, a Noncovalent Small Molecule Binder to Keap1 and Selective Activator of Nrf2 Signaling. J. Biol. Chem. 2015, 290, 28446–28455. [Google Scholar] [CrossRef]
  69. Unni, S.; Deshmukh, P.; Krishnappa, G.; Kommu, P.; Padmanabhan, B. Structural insights into the multiple binding modes of Dimethyl Fumarate (DMF) and its analogs to the Kelch domain of Keap1. FEBS J. 2021, 288, 1599–1613. [Google Scholar] [CrossRef]
  70. Ortet, P.C.; Muellers, S.N.; Viarengo-Baker, L.A.; Streu, K.; Szymczyna, B.R.; Beeler, A.B.; Allen, K.N.; Whitty, A. Recapitulating the Binding Affinity of Nrf2 for KEAP1 in a Cyclic Heptapeptide, Guided by NMR, X-ray Crystallography, and Machine Learning. J. Am. Chem. Soc. 2021, 143, 3779–3793. [Google Scholar] [CrossRef]
  71. Edelsbrunner, H. Weighted Alpha Shapes; Technical Paper of the Department of Computer Science of the University of Illinois at Urbana-Champaign; Urbana, Illinois 61810. Available online: https://pub.ista.ac.at/~edels/Papers/1992-R-08-WeightedAlphaShapes.pdf (accessed on 27 June 2023).
  72. Del Carpio, C.A.; Takahashi, Y.; Sasaki, S. A new approach to the automatic identification of candidates for ligand receptor sites in proteins: (I). Search for pocket regions. J. Mol. Graph. 1993, 11, 23–42. [Google Scholar] [CrossRef] [PubMed]
  73. Edelsbrunner, H.; Facello, M.; Fu, P.; Liang, J. Measuring Proteins and Voids in Proteins. In Proceedings of the 28th Hawaii International Conference on Systems Science, Wailea, HI, USA, 3–6 January 1995; Volume 5, pp. 256–264. [Google Scholar] [CrossRef]
  74. Goodford, P.J. A computational procedure for determining energetically favorable binding sites on biologically important macromolecules. J. Med. Chem. 1985, 28, 849–857. [Google Scholar] [CrossRef] [PubMed]
  75. Hendlich, M.; Rippmann, F.; Barnickel, G. LIGSITE: Automatic and efficient detection of potential small molecule-binding sites in proteins. J. Mol. Graph. Model. 1997, 15, 359–389. [Google Scholar] [CrossRef] [PubMed]
  76. Le Guilloux, V.; Schmidtke, P.; Tuffery, P. Fpocket: An open source platform for ligand pocket detection. BMC Bioinform. 2009, 10, 168. [Google Scholar] [CrossRef]
  77. Miranker, A.; Karplus, M. Functionality maps of binding sites: A multiple copy simultaneous search method. Proteins 1991, 11, 29–34. [Google Scholar] [CrossRef]
  78. Soga, S.; Shirai, H.; Kobori, M.; Hirayama, N. Use of amino acid composition to predict ligand-binding sites. J. Chem. Inf. Model. 2007, 47, 400–406. [Google Scholar] [CrossRef]
  79. Volkamer, A.; Griewel, A.; Grombacher, T.; Rarey, M. Analyzing the topology of active sites: On the prediction of pockets and subpockets. J. Chem. Inf. Model. 2010, 50, 2041–2052. [Google Scholar] [CrossRef]
Figure 1. Chemical structure of TBHQ.
Figure 1. Chemical structure of TBHQ.
Ijms 25 00475 g001
Figure 2. Homeostatic conditions of Nrf2–Keap1 and the proposed effect of TBHQ on the Nrf2–Keap1 pathway. Image created from BioRender.com.
Figure 2. Homeostatic conditions of Nrf2–Keap1 and the proposed effect of TBHQ on the Nrf2–Keap1 pathway. Image created from BioRender.com.
Ijms 25 00475 g002
Figure 3. Representative images of in situ livers of rats from the CCl4 and BDL models.
Figure 3. Representative images of in situ livers of rats from the CCl4 and BDL models.
Ijms 25 00475 g003
Figure 4. Representative micrographs of H&E-stained liver tissue sections from the CCl4 model. Photographs obtained from samples at different magnifications (5X, 10X, and 40X) show the differences and alterations in the tissue according to the different treatment groups.
Figure 4. Representative micrographs of H&E-stained liver tissue sections from the CCl4 model. Photographs obtained from samples at different magnifications (5X, 10X, and 40X) show the differences and alterations in the tissue according to the different treatment groups.
Ijms 25 00475 g004
Figure 5. Representative micrographs of liver sections stained with H&E from the BDL model. Photographs obtained from samples at different magnifications (5X, 10X, and 40X).
Figure 5. Representative micrographs of liver sections stained with H&E from the BDL model. Photographs obtained from samples at different magnifications (5X, 10X, and 40X).
Ijms 25 00475 g005
Figure 6. Evaluation of serum markers for liver damage. (a,d): ALT; (b,e): GGT; (c,f): ALP. CCl4 model (ac): Control, CCl4: CCl4 (1.6 g/kg, i.p.), CCl4+TBHQ: CCl4 (1.6 g/kg, i.p.) + TBHQ pretreatment and TBHQ groups BDL model; (df): SHAM: SHAM surgery; BDL: bile duct ligation; BDL+TBHQ and SHAM+TBHQ groups. Results were expressed as the mean ± SE. Statistically significant differences: (*) p < 0.05, (**) p < 0.01, (***), p < 0.001, and (****) p < 0.0001.
Figure 6. Evaluation of serum markers for liver damage. (a,d): ALT; (b,e): GGT; (c,f): ALP. CCl4 model (ac): Control, CCl4: CCl4 (1.6 g/kg, i.p.), CCl4+TBHQ: CCl4 (1.6 g/kg, i.p.) + TBHQ pretreatment and TBHQ groups BDL model; (df): SHAM: SHAM surgery; BDL: bile duct ligation; BDL+TBHQ and SHAM+TBHQ groups. Results were expressed as the mean ± SE. Statistically significant differences: (*) p < 0.05, (**) p < 0.01, (***), p < 0.001, and (****) p < 0.0001.
Ijms 25 00475 g006
Figure 7. Three-dimensional and two-dimensional interaction diagrams. Keap1 with (a) RA839, (b,c) MEF, and (d) the Kelch domain of Nrf2.
Figure 7. Three-dimensional and two-dimensional interaction diagrams. Keap1 with (a) RA839, (b,c) MEF, and (d) the Kelch domain of Nrf2.
Ijms 25 00475 g007
Figure 8. Common residues involved in the interaction present in each complex between the molecule and Nrf2. The MOE site-finding tool provides a high probability region filled with a set of dummy atoms for further docking experiments. Among the near residues are Arg380, Asp382, Ser363, Gly364, Tyr334, Gly603, Ser602, Tyr572, Ser555, and Tyr525.
Figure 8. Common residues involved in the interaction present in each complex between the molecule and Nrf2. The MOE site-finding tool provides a high probability region filled with a set of dummy atoms for further docking experiments. Among the near residues are Arg380, Asp382, Ser363, Gly364, Tyr334, Gly603, Ser602, Tyr572, Ser555, and Tyr525.
Ijms 25 00475 g008
Figure 9. Three-dimensional and two-dimensional interaction diagrams of Keap1 with TBHQ: (a) front, (b) side, and (c) 2D.
Figure 9. Three-dimensional and two-dimensional interaction diagrams of Keap1 with TBHQ: (a) front, (b) side, and (c) 2D.
Ijms 25 00475 g009
Figure 10. Three-dimensional interaction diagram of Keap1 with TBHQ allocated deep in the channel.
Figure 10. Three-dimensional interaction diagram of Keap1 with TBHQ allocated deep in the channel.
Ijms 25 00475 g010
Figure 11. In vivo experimental design to evaluate the ability of TBHQ to prevent rat mortality from a lethal dose of CCl4. Twelve rats were divided into two groups for the lethal toxicity assay of CCl4 (4 g/kg, i.p.): CCl4 (n = 6) and CCl4+TBHQ (n = 6). CCl4: carbon tetrachloride; TBHQ: Tert-butylhydroquinone; DMSO: dimethylsulfoxide (vehicle for TBHQ); i.p.: intraperitoneal route.
Figure 11. In vivo experimental design to evaluate the ability of TBHQ to prevent rat mortality from a lethal dose of CCl4. Twelve rats were divided into two groups for the lethal toxicity assay of CCl4 (4 g/kg, i.p.): CCl4 (n = 6) and CCl4+TBHQ (n = 6). CCl4: carbon tetrachloride; TBHQ: Tert-butylhydroquinone; DMSO: dimethylsulfoxide (vehicle for TBHQ); i.p.: intraperitoneal route.
Ijms 25 00475 g011
Figure 12. Experimental protocol to evaluate the hepatoprotective capacity of TBHQ in acute liver damage induced by a sublethal dose of CCl4. Twenty-four rats were divided into four groups for the sublethal toxicity test of CCl4 (1.6 g/kg, i.p.): Control (n = 6), CCl4 (n = 6), CCl4+TBHQ (n = 6), and TBHQ (n = 6). CCl4: carbon tetrachloride; TBHQ: Tert-butylhydroquinone; DMSO: dimethyl sulfoxide (vehicle for TBHQ); i.p.: intraperitoneal route.
Figure 12. Experimental protocol to evaluate the hepatoprotective capacity of TBHQ in acute liver damage induced by a sublethal dose of CCl4. Twenty-four rats were divided into four groups for the sublethal toxicity test of CCl4 (1.6 g/kg, i.p.): Control (n = 6), CCl4 (n = 6), CCl4+TBHQ (n = 6), and TBHQ (n = 6). CCl4: carbon tetrachloride; TBHQ: Tert-butylhydroquinone; DMSO: dimethyl sulfoxide (vehicle for TBHQ); i.p.: intraperitoneal route.
Ijms 25 00475 g012
Figure 13. Experimental protocol to evaluate the hepatoprotective effect of TBHQ on acute liver damage induced by extrahepatic cholestasis: (a) general protocol and (b) schematic representation of the surgical process of BDL (image created from BioRender.com). A total of 40 rats were divided into four groups: SHAM (n = 10), BDL (n = 10), BDL+TBHQ (n = 10), and SHAM+TBHQ (n = 10).
Figure 13. Experimental protocol to evaluate the hepatoprotective effect of TBHQ on acute liver damage induced by extrahepatic cholestasis: (a) general protocol and (b) schematic representation of the surgical process of BDL (image created from BioRender.com). A total of 40 rats were divided into four groups: SHAM (n = 10), BDL (n = 10), BDL+TBHQ (n = 10), and SHAM+TBHQ (n = 10).
Ijms 25 00475 g013
Figure 14. Schematic representation of the domains of Nrf2.
Figure 14. Schematic representation of the domains of Nrf2.
Ijms 25 00475 g014
Table 1. Mortality and survival of CCl4 and BDL experimental models.
Table 1. Mortality and survival of CCl4 and BDL experimental models.
Liver Damage ModelMortality
(n)%Survival %
Lethal dose of CCl4
CCl4 (4 g/kg, i.p.)6/61000
CCl4 (4 g/kg, i.p.) + TBHQ3/65050
Sublethal dose of CCl4
CCl4 (1.6 g/kg, i.p.)0/60100
CCl4 (1.6 g/kg, i.p.) + TBHQ0/60100
BDL experimental model
BDL5/105050
BDL+TBHQ5/102080
Table 2. Body and liver weight of the rats in hepatotoxicity model with sublethal dose of CCl4 and BDL model.
Table 2. Body and liver weight of the rats in hepatotoxicity model with sublethal dose of CCl4 and BDL model.
GroupBody Weight (g)Liver Weight (g)Liver/Body Weight
Control348.3 ± 4.7011.5 ± 0.650.033 ± 0.00169
CCl4350.8 ± 12.9816.2 ± 0.67 a0.046 ± 0.00194 ac
CCl4+TBHQ302.9 ± 8.26 ab15.21 ± 0.76 a0.050 ± 0.00178 ac
TBHQ370.3 ± 9.5313.92 ± 0.370.038 ± 0.00010
SHAM411.6 ± 3.3216.23 ± 0.0610.039 ± 0.00179
BDL373.38 ± 16.5515.36 ± 0.760.040 ± 0.00323
BDL+TBHQ366.9 ± 11.6214.48 ± 0.130.039 ± 0.00143
SHAM+TBHQ335.93 ± 12.4314.6 ± 0.490.043 ± 0.00056
Results were expressed as the mean ± SE. Statistically significant differences, p < 0.05: a significantly different from the Control group; b significantly different from the CCl4 group; c significantly different from the SHAM+TBHQ group.
Table 3. Putative protein targets for TBHQ in liver found in different databases.
Table 3. Putative protein targets for TBHQ in liver found in different databases.
Target Protein Name Protein IDTarget Location and/or Function
(Human Protein Atlas)
Target Search Platform
Nuclear factor erythroid 2-related factorNFE2L2 (Nrf2)Antioxidant
Liver—Metabolism
P
Nuclear factor-kappa B p105 subunitNFKB1NF-KB subunitSP
Transcription factor p65RELANF-KB subunitTN
Acetyl-CoA carboxylase 2ACACBCell type enhanced—HepatocytesSP
Estrogen receptorESR1Cell type enhanced—HepatocytesPM, S, TN
Endoplasmic reticulum-associated amyloid beta-peptide-binding proteinHSD17B10Cell type enhanced—HepatocytesP, SP
Dihydropteridine reductaseQDPRCell type enhanced—HepatocytesS
Nuclear receptor ROR gammaRORCCell type enhanced—HepatocytesP
Excitatory amino acid transporter 3SLC1A1Cell type enhanced—HepatocytesSP
TransthyretinTTRCell type enriched—HepatocytesPM, SP
Tyrosine-protein kinase receptor RETNRTNCell type enhanced—HepatocytesSP
Oxysterols receptor LXR-alphaNR1H3Cell type enhanced—HepatocytesTN
Glyceraldehyde-3-phosphate dehydrogenase, liver *GAPDHTissue enhanced—LiverST
Hydroxysteroid 17-beta-dehydrogenase 3HSD17B3Tissue enhanced—Liver TN
Pyruvate dehydrogenase kinase 4PDK4Cell type enriched—Liver–HepatocytesS
Aryl hydrocarbon receptorAHRLiver—MetabolismTN
Transient receptor potential cation channel subfamily V member 1TRPV1Liver—MetabolismTN
Aldo-keto reductase family 1 member C3AKR1C3Liver—Lipid metabolismPM
Monoamine oxidase AMAOALiver—Lipid metabolismSP, TN
Androgen receptorARLiver—Metabolism
Cell type enhanced—Hepatocytes
PM, TN
Coagulation factor XF10Liver—Metabolism
Cell type enhanced—Hepatocytes
PM
Monoamine oxidase BMAOBLiver—Metabolism
Cell type enhanced—Hepatocytes
TN
Bile acid receptorNR1H4Liver—Metabolism
Cell type enhanced—Hepatocytes
S
Cholinergic receptor nicotinic alpha 4 subunit (neuronal acetylcholine receptor)CHRNA4Liver—Metabolism
Cell type enhanced—Hepatocytes
SP
Cocaine esteraseCES2Liver—Lipid metabolism
Cell type enhanced—Hepatocytes
TN
Nuclear receptor subfamily 1 group I member 2 (pregnane X receptor)NR1I2Liver—Lipid metabolism
Cell type enhanced—Hepatocytes
SP, PM
Estrogen sulfotransferaseSULT1E1Liver—Lipid metabolism
Cell type enhanced—Hepatocytes
PM
Xanthine dehydrogenaseXDHLiver—Lipid metabolism
Cell type enhanced—Hepatocytes
Ph, SP, TN
Serum albuminALBTissue enriched—Liver
Liver—Hemostasis
Cell type enriched—Hepatocytes
PM, ST
Complement factor BCFBTissue enriched—Liver
Liver—Hemostasis
Cell type enhanced—Hepatocytes
PM
ProthrombinF2Liver—Hemostasis
Cell type enriched—Hepatocytes
PM
Hydroxysteroid 11-beta dehydrogenase 1HSD11B1Liver—Hemostasis and lipid metabolism
Cell type enriched—Hepatocytes
SP, TN
ButyrylcholinesteraseBCHELiver—Hemostasis and lipid metabolism
Cell type enhanced—Hepatocytes
Ch
Carbonic anhydrase 5A, mitochondrialCA5ALiver—Hemostasis and lipid metabolism
Cell type enriched—Hepatocytes
TN
Liver carboxylesterase 1 *CES1Liver—Hemostasis and lipid metabolism
Cell type enriched—Hepatocytes
TN
Cytochrome P450 1A2CYP1A2Liver—Hemostasis and lipid metabolism
Cell type enriched—Hepatocytes
TN
Cytochrome P450 2C9CYP2C9Liver—Hemostasis and lipid metabolism
Cell type enriched—Hepatocytes
P, TN
IgG receptor FcRn large subunit p51FCGRTLiver—Lipid metabolism
Cell type enhanced—Kupffer cells
SP
Arachidonate 5-lipoxygenaseALOX5Cell type enhanced—Kupffer cellsSP, ST, TN
C-C chemokine receptor type 2CCR2Cell type enhanced—Kupffer cellsSP
Tyrosine-protein kinase FGRFGRCell type enhanced—Kupffer cellsSP
Formyl peptide receptor 1FPR1Cell type enhanced—Kupffer cellsSP
Lipoxin A4 receptorFPR2Cell type enhanced—Kupffer cellsSP
G-protein coupled bile acid receptor 1GPBAR1Cell type enhanced—Kupffer cellsSP
Tyrosine-protein kinase SYKSYKCell type enhanced—Kupffer cellsPM
Hydroxysteroid17-beta-dehydrogenase 2HSD17B2Liver—ER transport pathway
Cell type enhanced—Hepatocytes
S
Endoplasmic reticulum aminopeptidase 1ERAP1Liver—ER transport pathwaySP
Tissue factor pathway inhibitorTFPITissue enhanced liver
Liver—ER transport pathway
SP
Tyrosine-protein kinase YESYES1Liver—ER transport pathwaySP
All those with the highest probability and that, according to the curated data from the Human Protein Atlas, had a liver-related tissue and/or cellular localization, and were specifically indicated (*) to be a liver protein. Ch—ChEMBL; PM—PharmMapper; Ph—Pharos; P—PPB; S—SEA; SP—Super-PRED; ST—SwissTargetPrediction; TN—TargetNet; ER—Endoplasmic reticulum.
Table 4. Potential binding site identified by SiteFinder. In bold, the amino acid residues that also participate in the interactions between Keap1 and the small molecules RA839 and MEF are highlighted.
Table 4. Potential binding site identified by SiteFinder. In bold, the amino acid residues that also participate in the interactions between Keap1 and the small molecules RA839 and MEF are highlighted.
SiteSizeHydSideResidues
Approximately the highest propensity for ligand binding192 alpha spheres (with the size of a carbon atom sphere)42 hydrophobic contact atoms in the receptor64 side-chain contact atoms in the receptorTYR334 SER363 GLY364 LEU365 ALA366 GLY367 CYS368 ASN382 ASN414 ARG415 ILE416 GLY417 VAL418 GLY419 VAL420 GLY462 VAL463 GLY464 VAL465 ALA466 VAL467 SER508 GLY509 ALA510 GLY511 VAL512 CYS513 VAL514 TYR525 GLN530 SER555 ALA556 LEU557 GLY558 ILE559 THR560 VAL561 TYR572 PHE577 SER602 GLY603 VAL604 GLY605 VAL606 ALA607 VAL608
Table 5. Interaction energies resulting from the molecular docking analysis.
Table 5. Interaction energies resulting from the molecular docking analysis.
LigandNef2/Nrf2MEFRA839TBHQ
Energy (kcal/mol)−10.4673−5.2186−6.9461−5.5491
Table 6. Nrf2 domains.
Table 6. Nrf2 domains.
DomainNameResidues/Amino Acids
Neh 1sMAF and DNA binding435–562
Neh 2Keap1 binding50–86
Neh 3Transactivation domain563–605
Neh 4Transactivation domain112–134
Neh 5Transactivation domain182–209
Neh 6p-TrCp binding338–388
Neh 7RARalfa binding210–316
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

Aldaba-Muruato, L.R.; Sánchez-Barbosa, S.; Rodríguez-Purata, V.H.; Cabrera-Cruz, G.; Rosales-Domínguez, E.; Martínez-Valentín, D.; Alarcón-López, Y.A.; Aguirre-Vidal, P.; Hernández-Serda, M.A.; Cárdenas-Granados, L.A.; et al. In Vivo and In Silico Studies of the Hepatoprotective Activity of Tert-Butylhydroquinone. Int. J. Mol. Sci. 2024, 25, 475. https://doi.org/10.3390/ijms25010475

AMA Style

Aldaba-Muruato LR, Sánchez-Barbosa S, Rodríguez-Purata VH, Cabrera-Cruz G, Rosales-Domínguez E, Martínez-Valentín D, Alarcón-López YA, Aguirre-Vidal P, Hernández-Serda MA, Cárdenas-Granados LA, et al. In Vivo and In Silico Studies of the Hepatoprotective Activity of Tert-Butylhydroquinone. International Journal of Molecular Sciences. 2024; 25(1):475. https://doi.org/10.3390/ijms25010475

Chicago/Turabian Style

Aldaba-Muruato, Liseth Rubi, Sandra Sánchez-Barbosa, Víctor Hugo Rodríguez-Purata, Georgina Cabrera-Cruz, Estefany Rosales-Domínguez, Daniela Martínez-Valentín, Yoshio Aldo Alarcón-López, Pablo Aguirre-Vidal, Manuel Alejandro Hernández-Serda, Luis Alfonso Cárdenas-Granados, and et al. 2024. "In Vivo and In Silico Studies of the Hepatoprotective Activity of Tert-Butylhydroquinone" International Journal of Molecular Sciences 25, no. 1: 475. https://doi.org/10.3390/ijms25010475

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