Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Biomedicine & Pharmacotherapy 103 (2018) 75–86

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

Betaine treatment protects liver through regulating mitochondrial function T


and counteracting oxidative stress in acute and chronic animal models of
hepatic injury

Reza Heidaria, , Hossein Niknahada,b, Ala Sadeghib, Hamidreza Mohammadib,
Vahid Ghanbarinejadb, Mohammad Mehdi Ommatia, Arghavan Hosseinib, Negar Azarpirac,
Forouzan Khodaeib, Omid Farshada,b, Elaheh Rashidib, Asma Siavashpourb, Asma Najibib,

Asrin Ahmadib, Akram Jamshidzadeha,b,
a
Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
b
Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
c
Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran

A R T I C LE I N FO A B S T R A C T

Keywords: Betaine is a derivative of the amino acid glycine widely investigated for its hepatoprotective properties against
Amino acid alcoholism. The protective properties of betaine in different other experimental models also have been docu-
ATP mented. On the other hand, the exact cellular mechanism of cytoprotection provided by betaine is obscure. The
Bile acid current study was designed to evaluate the hepatoprotective effects of betaine and its potential mechanisms of
Bioenergetics
hepatoprotection in two animal models of acute and chronic liver injury. Bile duct ligation (BDL) was used as a
Cholestasis
Liver injury
model of chronic liver injury and thioacetamide (TAA)-induced hepatotoxicity was applied as the acute liver
Oxidative stress injury model. Severe increase in serum markers of liver tissue damage along with significant liver tissue his-
topathological changes were evident in both acute and chronic models of hepatic injury. It was also found that
tissue markers of oxidative stress were significantly increased in BDL and TAA-treated animals. Moreover, liver
mitochondrial indices of functionality were deteriorated in both investigated models. Betaine supplementation
(10 and 50 mg/kg, i.p) ameliorated hepatic injury as judged by decreased liver tissue histopathological altera-
tions, a significant decrease in tissue markers of oxidative stress, and mitigation of serum biomarkers of hepa-
totoxicity. On the other hand, betaine (10 and 50 mg/kg, i.p) protected hepatocytes mitochondria in both
chronic and acute models of hepatotoxicity. These data indicate that the antioxidative and mitochondria reg-
ulating properties of betaine could play a primary role in its mechanisms of hepatoprotection.

1. Introduction mitochondrial dysfunction has also been reported in several human


pathological conditions and xenobiotics-induced cytotoxicity [14].
Betaine (Trimethylglycine) enters the human body through different Mitochondrial injury and inability to maintain sufficient cellular ATP
dietary sources [1,2]. Some physiological roles including osmor- level could lead to cell death [15]. Oxidative stress and mitochondrial
egulatory properties have been attributed to betaine [1,2]. This che- dysfunction are involved in the pathogenesis of liver injury in different
mical also participates in many critical biochemical pathways as a experimental models [10,16–19]. It has been revealed that TAA model
“methyl-donor” [1,2]. The hepatoprotective properties of betaine have of the acute liver injury is associated with severe oxidative stress and
repeatedly been mentioned in different experimental models [3–6]. mitochondrial dysfunction [20,21]. On the other hand, accumulation of
Betaine is also a well-known agent which protect the liver against al- the cytotoxic bile acid during bile duct obstruction is associated with
coholism [5,7,8]. mitochondrial injury and oxidative stress in the liver [22–25].
Previous investigations have provided compelling evidence that It has been found that betaine supplementation might blunt oxida-
oxidative stress and its associated events play a pivotal role in hepa- tive stress and its consequences in different experimental models
totoxicity with different etiologies [9–13]. On the other hand, [3,4,6,8,26]. Betaine also has been reported to prevent mitochondria-


Corresponding authors at: Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
E-mail addresses: rheidari@sums.ac.ir (R. Heidari), ajamshid@sums.ac.ir (A. Jamshidzadeh).

https://doi.org/10.1016/j.biopha.2018.04.010
Received 24 February 2018; Received in revised form 29 March 2018; Accepted 2 April 2018
0753-3322/ © 2018 Elsevier Masson SAS. All rights reserved.
R. Heidari et al. Biomedicine & Pharmacotherapy 103 (2018) 75–86

dependent cell death and apoptosis [7,27,28]. Hence, this chemical Betaine (50 mg/kg, i.p) + Thioacetamide.
might provide promising protective properties in animal models of Animals were anesthetized (Thiopental, 80 mg/kg, i.p) and their
hepatic injury. blood and liver samples were collected. Supportive therapy by admin-
Although the beneficial properties of betaine in different experi- istering 5% dextrose-containing 0.45% sodium chloride and 0.2% po-
mental models have been well-studied, the precise mechanism(s) of tassium chloride (2.5 ml/kg body weight, S.C), was given to avoid hy-
cytoprotection behind remained obscure [3,4,8]. The current study was poglycemia and renal failure [32]. Control animals (Vehicle-treated)
designed to evaluate the effect of betaine supplementation on liver received normal saline as the thioacetamide solvent.
mitochondrial function and oxidative stress in two experimental models
of acute (Thioacetamide; TAA treatment) and chronic (Bile duct liga- 2.5. Serum biochemistry
tion; BDL) liver injury. The data could help to clear the mechanism of
hepatoprotection provided by betaine as well as developing new ther- Animals were anesthetized (Thiopental, 50 mg/kg, i.p) and their
apeutic strategies against liver injury with different etiologies. blood, liver, and kidney samples were collected. Blood was collected
from the abdominal aorta, transferred to standard tubes
2. Materials and methods (Improvacuter®; gel and clot activator-coated tubes; Guangzhou, China)
and centrifuged (3000 g, 10 min, 4 °C) to prepare serum. Mindray BS-
2.1. Chemicals 200® autoanalyzer (Mindray chemistry analyzers for low-volume la-
boratories, Guangzhou, China) was used to analyze serum biochem-
4,2-Hydroxyethyl,1-piperazineethanesulfonic acid (HEPES), 3-(N- istry. Standard kits (Pars Azmun®, Tehran, Iran) were employed to as-
morpholino)propanesulfonic acid (MOPS), Dimethyl sulfoxide (DMSO), sess serum gamma-glutamyl transpeptidase (γ-GT), alkaline
D-mannitol, Bovine serum albumin (BSA), Thiobarbituric acid (TBA), 3- phosphatase (ALP), Creatinine (Cr), glucose, phosphate, calcium, uric
[4,5dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT), di- acid and blood urea nitrogen (BUN) [33].
thiobis-2nitrobenzoic acid (DTNB), Glutathione (GSH), 2′,7′-
Dichlorofluorescein diacetate (DCFH-DA), Betaine (Tri-methyl glycine), 2.6. Reactive oxygen species formation
Malondialdehyde (MDA), Sucrose, n-Propanol, n-Butanol, Sodium ci-
trate, Potassium chloride, di-Sodium hydrogen phosphate (Na2HPO4), Reactive oxygen species (ROS) in the liver was estimated as pre-
Sodium succinate, Glacial acetic acid, Magnesium chloride, viously described [20,34,35]. Briefly, liver tissue (500 mg) was homo-
Dithiothreitol, Rhodamine123 (Rh 123), Coomassie brilliant blue, genized in 5 ml of ice-cooled Tris-HCl buffer (40 mM, pH = 7.4, 4 °C).
Ethyleneglycol-bis (2-aminoethylether)-N,N,N′,N′-tetraacetic acid Samples of the resulted tissue homogenate (100 μL) were mixed with
(EGTA), and Ethylenediaminetetraacetic acid (EDTA) were purchased Tris-HCl buffer (pH = 7.4, 1 mL) and 2′, 7′dichlorofluorescein diacetate
from Sigma Chemical Co. (St. Louis, MO, USA). Sodium bicarbonate, (DCF-DA; Final concentration 10 μM). The mixture was incubated in the
Trichloroacetic acid (TCA), Sodium acetate, and Hydroxymethyl amino dark (37 °C; 15 min). Finally, the fluorescence intensity of the samples
methane-hydrochloride (Tris-HCl) were purchased from Merck was assessed using a FLUOstar Omega® multi-functional microplate
(Darmstadt, Germany). All salts for preparing buffer solutions were of reader with λ excitation = 485 nm and λ emission = 525 nm [34,36].
analytical grade and prepared from Merck (Darmstadt, Germany).
2.7. Lipid peroxidation
2.2. Animals
The thiobarbituric acid reactive substances (TBARS) were measured
Male Sprague Dawley rats (200–250 g) were obtained from Animal as an index of lipid peroxidation in the liver tissue [20]. Briefly, the
Breeding Center of Shiraz University of Medical Sciences, Shiraz Iran. reaction mixture was consisted of 500 μL of liver tissue homogenate
Rats were housed in cages on wood-chip bedding at a temperature of (10% w/v in KCl, 1.15% w/v), 1 mL of thiobarbituric acid (0.375%, w/
23 ± 2 °C and relative humidity ≈40%. Animals had free access to v), and 3 mL of phosphoric acid (1% w/v, pH = 2) [37]. Samples were
food (Behparvar®, Tehran, Iran) and tap water. Animals were handled mixed well and heated (100 °C). After the incubation period (45 min),
according to the guidelines approved by a local ethics committee at the mixture was cooled, and then 2 mL of n-butanol was added. Samples
Shiraz University of Medical Sciences, Shiraz, Iran (95-01-36-12046). were vigorously mixed and centrifuged (10,000 g for 10 min) [38].
Finally, the absorbance of developed color in n-butanol phase was
2.3. Animal surgery and bile duct ligation (BDL) as the model of chronic measured at λ = 532 nm using an Ultrospec 2000®UV spectro-
liver injury photometer (Pharmacia Biotech, Uppsala, Sweden) [20].

Animals were anesthetized (A mixture of Ketamine 80 mg/kg and 2.8. Hepatic glutathione content
Xylazine 10 mg/kg, i.p), a midline incision was made, and the common
bile duct was localized, doubly ligated, and cut between these ligatures Liver samples (500 mg) were homogenized in 8 ml of ice-cooled
[29]. The sham operation comprised laparotomy and bile duct identi- (4 °C) EDTA solution (0.04 M). Then, 5 mL of the prepared homogenate
fication and manipulation without ligation. The treatments in chronic were added to 4 mL of distilled water (4 °C) and 1 mL of trichloroacetic
liver injury model were as follow: 1) Sham-operated; 2) BDL; 3) acid (50% w/v; 4 °C). The mixture was vortexed and centrifuged
BDL + Betaine (10 mg/kg/day, i.p); 4) BDL + Betaine (50 mg/kg/day, (10,000 g, 4 °C, 15 min) [37]. Then, 2 mL of the supernatant was mixed
i.p). with 4 mL of Tris-HCl buffer (40 mM, pH = 8.9), and 100 μl of DTNB
(10 mM in methanol) [38,39]. The absorbance of the developed color
2.4. Animal model of acute liver injury was measured at λ = 412 nm using an Ultrospec 2000®UV spectro-
photometer (Pharmacia Biotech, Uppsala, Sweden) [20].
Thioacetamide-induced hepatotoxicity is extensively used as an
animal model of acute liver injury [30]. In the current investigation, 2.9. Ferric reducing antioxidant power (FRAP) of liver tissue
thioacetamide-induced liver injury was achieved by i.p injection of
thioacetamide (200 mg/kg) to rats [31]. Betaine (10 and 50 mg/kg, i.p) The FRAP assay measures the formation of a blue colored Fe2+-
was administered for three consecutive days before thioacetamide tripyridyl-triazine compound from the colorless oxidized Fe3+ form by
challenge. The treatments were as follow: 1) Control (Vehicle-treated); the action of electron-donating antioxidants [40]. In the current study,
2) Thioacetamide; 3) Betaine (10 mg/kg, i.p) + Thioacetamide; 4) the working FRAP reagent was prepared by mixing 10 volumes of

76
R. Heidari et al. Biomedicine & Pharmacotherapy 103 (2018) 75–86

Table 1
Serum level of liver injury biomarkers in bile duct ligated (BDL) and thioacetamide (TAA)-treated rats.
Treatment Serum ALT (U/l) Serum AST (U/l) Serum LDH (U/l) Serum ALP (U/l) Serum Bilirubin (mg/dl) Serum γGT (IU/l) Serum Albumin (g/dl)

Control 33 ± 3 92 ± 4 433 ± 25 1055 ± 115 0.08 ± 0.006 39 ± 7 2.98 ± 0.21


BDL 342 ± 28* 220 ± 12* 623 ± 71* 3814 ± 115* 11.8 ± 0.60* 265 ± 16* 1.57 ± 0.41*
BDL + Betaine 10 mg/kg 267 ± 14 a 154 ± 10 a 687 ± 26 2548 ± 173 9.2 ± 1.40 272 ± 25 2.42 ± 0.12a
BDL + Betaine 50 mg/kg 190 ± 11 a 119 ± 11 a 447 ± 16 a 3335 ± 308 9.5 ± 1.14 190 ± 22 2.3 ± 0.05
TAA 1103 ± 144* 1532 ± 133* 1043 ± 115* 2553 ± 514* 2.3 ± 0.2* 44 ± 7 1.9 ± 0.07*
TAA + Betaine 10 mg/kg 723 ± 58 b 320 ± 40 b 660 ± 69 b 2409 ± 184 1.0 ± 0.07 b 30 ± 4 2.4 ± 0.1b
TAA + Betaine 50 mg/kg 560 ± 55 b 377 ± 62 b 402 ± 70 b 2643 ± 240 0.67 ± 0.15 b 59 ± 8 2.6 ± 0.08 b

Data are given as Mean ± SEM (n = 6).


* Indicates significantly different as compared with control group (P < 0.001).
a
Indicates significantly different as compared with BDL group (P < 0.01).
b
Indicates significantly different as compared with TAA group (P < 0.01).

Fig. 1. Markers of oxidative stress in the liver tissue of cirrhotic rats. BDL: Bile duct ligation.
Data are given as Mean ± SEM (n = 6).
***Indicates significantly different as compared with sham-operated group (P < 0.001).
a
Indicates significantly different as compared with BDL group (P < 0.01).
ns: not significant.

acetate buffer (300 mmol/L, pH = 3.6), with 1 vol of TPTZ (10 mmol/L spectrophotometer (Pharmacia Biotech, Uppsala, Sweden) [35,41].
in 40 mmol/L hydrochloric acid) and 1 vol of ferric chloride (20 mmol/
L). All solutions were freshly-prepared and stored in the dark (4 °C). 2.10. Liver hydroxyproline level
Liver tissue (500 mg) was homogenized in an ice-cooled Tris-HCl buffer
(250 mM Tris-HCl, 200 mM sucrose and 5 mM DTT, pH = 7.4, 4 °C) Liver hydroxyproline content was assessed as an index of tissue fi-
[39]. Then, 50 μL of tissue homogenate and 150 μL of deionized water brosis. Briefly, 500 μl of the tissue homogenate (20% w/v in phosphate
was added to 1.5 mL of the FRAP reagent [21]. The reaction mixture buffered saline; PBS; pH = 7.4) was digested in 1 ml of hydrochloric
was incubated at 37 °C for 5 min. Finally, the absorbance of developed acid (6 N) at 120 °C for at least 12 h. An aliquot of the digested
color was measured at λ = 595 nm using an Ultrospec 2000® UV homogenate (25 μl) was added to 25 μl of citrate-acetate buffer

77
R. Heidari et al. Biomedicine & Pharmacotherapy 103 (2018) 75–86

Fig. 2. Effect of betaine treatment on the liver tissue markers of oxidative stress in an animal model of acute liver injury. TAA: Thioacetamide.
Data are given as Mean ± SEM (n = 6).
***Indicates significantly different as compared with control group (P < 0.001).
a
Indicates significantly different as compared with TAA group (P < 0.01).
ns: not significant.

(pH = 6). Afterward, 500 μl of chloramines-t-solution (56 mM) was fresh buffer medium. As mentioned, all procedures were performed at
added, and the mixture was left at room temperature for 20 min. Then, 0–4 °C or on ice to minimize mitochondrial damage during the mi-
500 μl of freshly-prepared Ehrlich's reagent (15 g of p-dimethyl amino tochondria isolation process [43].
benzaldehyde in n-propanol/perchloric acid; 2: 1 v: v) was added, and
the mixture was incubated at 65 °C for 15 min [42]. After cooling, the 2.12. Mitochondrial dehydrogenases activity (MTT assay)
intensity of developed color was measured at λ = 550 nm (Ultrospec
2000® UV spectrophotometer; Pharmacia Biotech, Uppsala, Sweden) The 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide
[20]. (MTT) assay was applied as a colorimetric method for determination of
mitochondrial succinate dehydrogenase activity in isolated mice liver
2.11. Liver mitochondria isolation mitochondria [39,45]. Mitochondrial suspension in a buffer containing
320 mM sucrose, 1 mM EDTA, and 10 mM Tris-HCl; pH = 7.4, was in-
Rat liver mitochondria were isolated based on a previously de- cubated with MTT (0.4% w: v; 37 °C for 30 min in the dark). The pro-
scribed method [43]. Briefly, animals were anesthetized (Thiopental duct of purple formazan crystals was dissolved in 1 mL dimethyl sulf-
80 mg/kg, i.p) and their liver was excised and washed with ice-cold oxide (DMSO). Then, 100 μl of dissolved formazan was added to 96 well
saline (sodium chloride 0.9%) [43,44]. The liver was homogenized in plate, and the optical density (OD) at λ = 570 nm was measured with
isolation buffer (220 mM sucrose, 70 mM mannitol, 0.5 mM ethylene an EPOCH plate reader (BioTek Instruments, Highland Park, USA) [46].
glycol-bis(2-aminoethyl ether)-N,N,N´,N´-tetraacetic acid (EGTA),
2 mM N-(2-hydroxyethyl) piperazine-N,N´-(2-ethane sulfonic acid) 2.13. Reactive oxygen species (ROS) in isolated liver mitochondria
(HEPES), 0.1% essentially fatty acid-free bovine serum albumin;
pH = 7.4) at a 10:1 buffer to tissue (v/w) ratio [43]. Afterward, tissue The fluorescent probe DCFH-DA was used to assess mitochondrial
homogenate was centrifuged at 1000 × g for 10 min at 4 °C to remove ROS [43,47]. Briefly, isolated liver mitochondria were placed in re-
intact cells and nuclei. The supernatants were further centrifuged at spiration buffer (125 mM sucrose, 65 mM KCl, 10 mM HEPES, 5 mM
15,000 × g (4 °C for 10 min) to precipitate the heavy membrane frac- Sodium succinate, 20 μM Ca2+, pH = 7.2) [22,43]. Following this step,
tions (mitochondria). This step was repeated at least three times using DCFH-DA was added (Final concentration, 10 μM) to mitochondria and

78
R. Heidari et al. Biomedicine & Pharmacotherapy 103 (2018) 75–86

Fig. 3. Mitochondrial indices of functionality in bile duct ligated (BDL) rats.


Data are given as Mean ± SEM (n = 6).
***Indicates significantly different as compared with sham-operated group (P < 0.001).
a
Indicates significantly different as compared with BDL group (P < 0.01).
ns: not significant.

then incubated for 30 min (37 °C in the dark). Then, the fluorescence experimental groups and reported as maximal mitochondrial swelling
intensity of DCF was measured using a FLUOstar Omega® multi- amplitude [43].
functional microplate reader (λ excitation = 485 nm and λ emission
= 525 nm) [43]. 2.16. Mitochondrial ATP content

2.14. Mitochondrial depolarization A luciferase-luciferin-based kit (Enliten® from Promega Corp.,


Madison, WI, USA) was used to assess mitochondrial ATP content
Mitochondrial uptake of the cationic fluorescent dye, rhodamine [21,52]. Samples and buffer solutions were prepared based on the kit
123, has been used for the estimation of mitochondrial depolarization instructions. Briefly, 500 μL of mitochondrial samples (1 mg protein/
[43,48]. For this purpose, the mitochondrial fractions (0.5 mg protein/ mL) were treated with 200 μL of ice-cooled TCA solution (0.3% w: v in
mL) were incubated with 10 μM of rhodamine 123 in a buffer con- double distilled water) and centrifuged (15,000 g, 15 min). Afterward,
taining 125 mM sucrose, 65 mM KCl, 10 mM HEPES, dissolved in 100 μL of the supernatant was treated with 100 μL of ATP kit content,
double distilled water, pH = 7.2 (30 min, 37 °C, in the dark). Samples and the luminescence intensity of samples was measured λ = 560 nm
were centrifuged (15,000 g, 10 min, 4 °C) and the fluorescence intensity using a FLUOstar Omega® multi-functional microplate reader. For
of supernatant was monitored using a FLUOstar Omega® multi- standardization of data, samples protein concentrations were de-
functional microplate reader at the excitation and emission wavelength termined using the Bradford method [53].
of λ = 485 nm and λ = 525 nm, respectively [43,49].
2.17. Statistical analysis
2.15. Mitochondrial swelling assay
Data are given as the Mean ± SEM. Data comparison was per-
Mitochondrial swelling was measured based on the light scattering formed by the one-way analysis of variance (ANOVA) with Tukey’s
method as previously described [43,50]. Briefly, the mitochondria multiple comparison test as the post hoc. Differences were considered
(0.5 mg protein/ml) were suspended in swelling buffer (125 mM su- statistically significant when P < 0.05.
crose, 65 mM KCl, 10 mM Hepes-KOH, 20 μM Ca2+, pH = 7.2). Light
absorbance at λ = 540 nm was monitored (Constant temperature of 3. Results
30 °C) [43,51]. It is accepted that a decreased light absorbance is con-
sistent with an increase in mitochondrial volume. Hence, as mi- Severe tissue damage was evident in both acute and chronic models
tochondria are more swelled, the differences between light absorbance of hepatotoxicity as judged by the significant increase in serum bio-
of two-time points are higher. The differences between the absorbance markers of liver injury (Table 1). It was found that betaine treatment
of samples were assessed (ΔOD540 nm) and compared in different (10 and 50 mg/kg, i.p) mitigated serum markers of hepatotoxicity

79
R. Heidari et al. Biomedicine & Pharmacotherapy 103 (2018) 75–86

Fig. 4. Mitochondrial dysfunction in the liver of thioacetamide (TAA)-treated rats.


Data are given as Mean ± SEM (n = 6).
***Indicates significantly different as compared with control group (P < 0.001).
a
Indicates significantly different as compared with TAA group (P < 0.01).

(Table 1). On the other hand, betaine supplementation (10 and 50 mg/ A significant decrease in mitochondrial function was also detected
kg, i.p) did not significantly change some markers of liver and bile duct in the liver of TAA-treated rats (Fig. 4). On the other hand, it was found
injury (e.g., bilirubin, γ-GT, and ALP) (Table 1). that betaine treatment (10 and 50 mg/kg, i.p) prevented TAA-induced
The markers of oxidative stress were significantly increased in cir- mitochondrial injury (Fig. 4). The mitochondrial protecting properties
rhotic BDL rats (Fig. 1). Severe ROS formation, along with an increase of betaine seem to be dose-dependent at the mitochondrial level in the
in liver tissue lipid peroxidation was detected in BDL group (Fig. 1). animal model of acute liver injury (Fig. 4).
Moreover, liver glutathione reservoirs were depleted, and tissue anti- Liver tissue histopathological changes were evident as severe ne-
oxidant capacity was significantly decreased in BDL rats (Fig. 1). It was crosis, inflammation, ballooning degeneration, and fatty changes in
found that betaine supplementation (10 and 50 mg/kg, i.p) alleviated BDL animals (Fig. 5 and Table 2). On the other hand, significant tissue
liver tissue markers of oxidative stress in BDL animals (Fig. 1). It is fibrosis and collagen deposition were detected in BDL rats (Fig. 6 and
noteworthy to mention that the effect of betaine on oxidative stress Table 2). It was found that betaine administration in both doses of 10
biomarkers was not dose-dependent in cirrhotic BDL animals in the and 50 mg/kg, alleviated liver tissue histopathological alterations in
current study (Fig. 1). cirrhotic animals (Figs. 5 and 6, Table 2).
Oxidative stress markers were significantly changed in TAA-treated Significant liver tissue necrosis, inflammation, and fatty changes
rats as the animal model of acute liver injury (Fig. 2). It was found that were also detected in TAA-treated (Fig. 7 and Table 2). It was found
betaine treatment (10 and 50 mg/kg, i.p) abated markers of oxidative that betaine (10 and 0 mg/kg, i.p) significantly decreased histopatho-
stress in TAA-treated animals (Fig. 2). The higher dose of betaine logical alterations in TAA-induced hepatotoxicity (Fig. 7 and Table 2).
(50 mg/kg, i.p) seems to have more significant effect against oxidative
stress in comparison with the lower dose in TAA model of acute liver
injury (Fig. 2). 4. Discussion
It was found that bile duct obstruction leads to significant dete-
rioration in the liver mitochondrial function (Fig. 3). Mitochondrial The current investigation was designed to evaluate the potential
ATP content and dehydrogenases activity were significantly decreased mechanisms of hepatoprotection provided by betaine in two experi-
where mitochondrial ROS formation and swelling were increased in mental models of acute and chronic liver injury. It was found that be-
BDL group (Fig. 3). Moreover, the significant collapse of mitochondrial taine supplementation (10 and 50 mg/kg) significantly decreased
membrane potential was detected in the liver mitochondria isolated markers of oxidative stress and efficiently preserved mitochondrial
from BDL animals (Fig. 3). It was found that betaine (10 and 50 mg/kg, function and energy metabolism in both acute and chronic hepato-
i.p) preserved mitochondrial indices of functionality in cirrhotic rats toxicity models. Hence, the mitochondrial protection and antioxidative
(Fig. 3). The effect of betaine on liver mitochondrial function was not properties could serve as fundamental mechanisms for the hepatopro-
dose-dependent in cirrhotic animals (Fig. 3). tective effects of betaine.
Previous investigations have provided compelling evidence that

80
R. Heidari et al. Biomedicine & Pharmacotherapy 103 (2018) 75–86

Fig. 5. Liver tissue histopathological changes


(H&E stain) in cirrhotic animals. BDL: Bile duct
ligated.
Severe tissue histopathological changes in-
cluding necrosis, inflammation, ballooning
degeneration, and fatty changes were detected
in the liver of BDL animals (Table 2). Betaine
supplementation (10 and 50 mg/kg, i.p) alle-
viated BDL-associated liver injury in cirrhotic
rats (Table 2).

Table 2
Effect of betaine supplementation on the liver histopathological alterations in acute and chronic models of liver injury.
Pathological assessment Confluent Necrosis Focal Necrosis Portal Inflammation InterfaceHepatitis Ballooning Total Ishak stage of liver
Treatment degeneration grade fibrosis

Control (vehicle-treated rats) 0 0 0 0 0 0 –


Bile duct ligated (BDL) rats 4 3 3 2 1 13 4
BDL + Betaine 10 mg/kg 0 0 2 1 1 4 2
BDL + Betaine 50 mg/kg 0 0 1 1 1 3 1
TAA 3 0 1 0 2 9 –
TAA + Betaine 10 mg/kg 2 0 0 0 1 7 –
TAA + Betaine 50 mg/kg 1 0 0 0 1 4 –

BDL: Bile duct ligation; TAA: Thioacetamide.

oxidative stress and its consequences play a critical role in different development of tissue fibrosis [64]. Cellular mitochondria dysfunction
liver diseases [9–13,54]. The crucial role of oxidative/nitrosative stress and its associated events play a critical role in the cell death process
in the pathogenesis of acute and chronic models of hepatic injury has [65–68]. Mitochondrial dysfunction also involved in the mechanism of
also been widely investigated [9,55,56]. The role of oxidative stress in a wide range of xenobiotics-induced liver injury [11,16,18–20,69,70].
the mechanism of liver injury and fibrogenesis also has largely been Hence, targeting this vital organelle could serve as a crucial pharma-
revealed [9,12,54]. Hence, antioxidant supplementation might be of cological point of intervention.
value against these complications. Betaine is abundantly found in daily food intake [1,2]. This che-
It has been found that the accumulation of cytotoxic bile acids in the mical is also endogenously synthesized in cellular mitochondria by the
rat model of BDL-induced cholestasis/cirrhosis is a well-documented oxidative metabolism of choline [1,2]. Betaine is then partially trans-
pathogenic factor associated with vast tissue injury [23,25,57–62]. ported from mitochondria to the cytoplasm [1,2]. On the other hand,
Hydrophobic bile acids are detergents which severely disrupt bio- betaine transporters are predominantly expressed in the liver which is
membrane lipids, and defect the nature and function of cellular proteins responsible for betaine accumulation in hepatocytes [2,71]. The hepa-
[25,58–62]. Changes in the liver tissue antioxidant status has also been toprotective properties of betaine have been shown against different
documented in acute and chronic animal models of hepatotoxicity [63]. complications including fatty liver, alcoholism–associated liver injury,
In line with previous studies, the data obtained in the current in- tissue fibrosis, and cellular necrosis [5–7,26,72–74]. However, the
vestigation mentioned the occurrence of oxidative stress and its asso- precise cellular mechanisms behind betaine cytoprotection remain
ciated complications in liver tissue of acute and chronic models of liver mostly unknown.
injury (Fig. 1 and 2). All these data could mention the crucial role of The antioxidant property of betaine is the dominant mechanism
antioxidant therapy. On the other hand, increasing evidence also sug- proposed for the cytoprotection provided by this chemical
gest that mitochondrial dysfunction can also contribute to the [5,72,75–79]. The role of betaine against oxidative stress and its

81
R. Heidari et al. Biomedicine & Pharmacotherapy 103 (2018) 75–86

Fig. 6. Effect of betaine supplementation on the liver tissue fibrosis and hydroxyproline content in cirrhotic rats (Masson Trichrome stain). BDL: Bile duct ligated.
Liver tissue score of fibrosis (Blue color) is given in Table 2. Data for liver hydroxyproline content and collagen deposition are given as Mean ± SEM (n = 6).
***Indicates significantly different as compared with control (P < 0.001).a Indicates significantly different as compared with BDL group (P < 0.01). ns: not
significant.

consequences has repeatedly been mentioned in different experimental in the reconstitution of damaged biological targets (e.g., membrane
models [5,72,75–79]. The effects of betaine on enzymatic and non- phospholipids, DNA) [84,85]. SAM also is essential for biomembrane
enzymatic cellular antioxidants have also been reported in previous lipids biosynthesis [85,86]. Hence, improving the biomembrane
investigations [5,72,75–79]. In the current study, we found that betaine synthesis might play a crucial role in the cytoprotective properties of
treatment efficiently abated oxidative stress and its associated compli- betaine. These data mention that a significant part of hepatoprotection
cations in both acute and chronic liver injury. provided by betaine might be associated with SAM production in the
Previous investigations also have shown that in addition to the liver and sustaining the function of the vital cell organelles such as
antioxidative properties, betaine serves as an ideal osmolyte in plant or mitochondria. SAM is also known as a radical scavenger and a chemical
animal cells [1,28,80,81]. The osmoprotective function of betaine has which participates in boosting cellular antioxidant defense mechanisms
also been reported in different biological systems [2,80]. Although it [5,84,85,87–89]. Therefore, a part of the antioxidant properties of be-
has not been evaluated in the current study, the osmoregulatory taine in the liver tissue might be attributed to its SAM metabolite.
properties of betaine might protect hepatocytes in the extreme condi- Betaine also efficiently alleviated liver fibrosis in different experi-
tions of bile duct obstruction or acute liver injury. mental models [72,79,90]. In accordance with these studies, we found
Some investigations revealed that betaine increases hepatocytes S- that betaine treatment of BDL animals significantly alleviated liver
Adenosyl methionine (SAM) level [82,83]. SAM plays an essential role tissue collagen deposition and hydroxyproline content (Fig. 6). The

82
R. Heidari et al. Biomedicine & Pharmacotherapy 103 (2018) 75–86

Fig. 7. Effect of betaine supplementation on


liver tissue histopathology in the animal model
of acute liver injury (H&E stain). TAA:
Thioacetamide.
Liver tissue histopathological changes in
thioacetamide-treated group revealed as in-
flammation, necrosis, and ballooning degen-
eration. Betaine supplementation (10 and
50 mg/kg respectively) mitigated thioaceta-
mide hepatotoxicity. The grade of histopatho-
logical changes in the acute model of hepatic
injury is given in Table 2.

antifibrotic properties of betaine could be mediated through a range of mitochondrial function and decreased cellular ATP content could also
mechanisms. As mentioned, oxidative/nitrosative stress and its asso- impair the cellular repairing processes and tissue regeneration. In the
ciated complications play a crucial role in the initiation and propaga- current study, we found that mitochondrial ATP content was higher in
tion of tissue fibrosis [9,12,54]. Hence, the antioxidative properties and betaine-treated groups in animal models of acute and chronic liver in-
modulation of cellular redox environment (e.g., by boosting cellular jury (Fig. 3 and 4). Therefore, sufficient ATP might be available for liver
antioxidant defense mechanisms) could play a significant role in the tissue regeneration in betaine-treated animals.
antifibrotic properties of betaine [5]. Hydrophobic bile acids are well-known mitochondrial toxins which
The role of inflammatory cells and mediators in the progression of their accumulation might be a causative factor for oxidative stress and
liver injury and tissue fibrosis has been well-documented in previous damage to hepatocytes during cholestasis/cirrhosis [9,23–25,62,104].
models of hepatotoxicity [57,91,92]. Interestingly, it has been revealed These chemicals severely affect mitochondrial function and impair
that betaine could abate inflammatory responses [76,93,94]. Although energy metabolism [9,23–25,62,104]. Bile acids induce mitochondrial
not evaluated in the current study, the anti-inflammatory properties of permeability transition, dissipate mitochondrial membrane potential,
betaine might play a role in its hepatoprotective properties especially in and finally cause the release of cell death mediators from mitochondria
chronic liver injury and cirrhosis. [9,23–25,62,104]. The inhibitory effects of bile acids on mitochondrial
The effect of betaine on tissue fibrosis also has been revealed in respiratory complexes also has been revealed [9,23–25,62,104]. On the
previous studies [72,95]. Stellate cells play a crucial role in liver fi- other hand, the relevance of mitochondrial dysfunction to the hepatic
brogenesis [96,97]. Therefore, the inhibitory effects of betaine on injury in animal models of cholestasis also has been mentioned in
stellate cells might also play a role in its mechanisms of hepatopro- previous studies [9,23–25,62,104]. Hence, preserving normal mi-
tection and anti-fibrogenesis. The anti-inflammatory effects of betaine tochondrial function could serve as a potential therapeutic point of
might also provide another possible explanation for its hepatoprotec- intervention during cholestasis. Our results showed the same pattern
tive properties in acute or chronic liver injury. Hence, the effect of and revealed significant impairment of mitochondrial function in an-
betaine on inflammatory cytokines and its relevance to the hepato- imal models of chronic or acute liver injury (Fig. 3 and 4). It was found
protective properties of this chemical deserve further investigations. that betaine supplementation preserved mitochondrial indices of func-
Interestingly, it has been mentioned that betaine supplementation tionality both in acute or chronic animal models of liver injury (Fig. 3
could prevent liver mitochondrial morphological alterations in rats and 4). These data mention the mitochondrial protection as a funda-
[98]. Recently, the direct effect of betaine on mitochondrial energy mental mechanism for the hepatoprotection provided by betaine. On
metabolism also has been reported in an in vitro system [99]. The the other hand, oral administration of betaine also prevented ROS
protective effects of betaine on mitochondrial function and mitochon- formation and lipid peroxidation. Moreover, betaine preserved liver
dria-mediated cell death process in other experimental models also tissue glutathione content and antioxidant capacity (Fig. 1 and 2).
have been revealed [28,100–103]. All these data indicate that mi- As mentioned, the effect of betaine on oxidative stress and its con-
tochondrial protecting properties of betaine could play a role in its sequences has been mentioned in several investigations [5,8,72,76]. On
mechanism of cytoprotection. On the other hand, the impairment of the other hand, oxidative stress and mitochondrial dysfunction are two

83
R. Heidari et al. Biomedicine & Pharmacotherapy 103 (2018) 75–86

mechanistically related events [105]. Hence, mitochondrial protecting Pharm. Bull. 5 (1) (2015) 1–11.
properties of betaine might also play a role in its ability to alleviate [18] D. Pessayre, B. Fromenty, A. Berson, M.-A. Robin, P. Lettéron, R. Moreau,
A. Mansouri, Central role of mitochondria in drug-induced liver injury, Drug
oxidative stress in the liver. Metab. Rev. 44 (1) (2012) 34–87.
In the current study, the insignificant changes in some markers of [19] S. Russmann, G.A. Kullak-Ublick, I. Grattagliano, Current concepts of mechanisms
the bile duct and liver injury (e.g., ALP, γ-GT, and bilirubin) upon be- in drug-induced hepatotoxicity, Curr. Med. Chem. 16 (23) (2009) 3041–3053.
[20] R. Heidari, H. Babaei, M.A. Eghbal, Amodiaquine-induced toxicity in isolated rat
taine treatment (Table 1) might be due to the permanent nature of bile hepatocytes and the cytoprotective effects of taurine and/or N-acetyl cysteine,
duct obstruction in BDL model. Res. Pharm. Sci. 9 (2) (2014) 97–105.
The data presented in the current study suggest that betaine might [21] A. Jamshidzadeh, R. Heidari, M. Abasvali, M. Zarei, M.M. Ommati, N. Abdoli,
F. Khodaei, Y. Yeganeh, F. Jafari, A. Zarei, Z. Latifpour, E. Mardani, N. Azarpira,
provide hepatoprotection through regulating mitochondrial function, B. Asadi, A. Najibi, Taurine treatment preserves brain and liver mitochondrial
membrane stabilizing action, and antioxidative effects in animal models function in a rat model of fulminant hepatic failure and hyperammonemia,
of acute or chronic liver injury. Further investigations should be per- Biomed. Pharmacother. 86 (2017) 514–520.
[22] R. Heidari, V. Ghanbarinejad, H. Mohammadi, A. Ahmadi, M.M. Ommati,
formed to clarify the effects of betaine on tricarboxylic acid cycle (TCA;
N. Abdoli, F. Aghaei, A. Esfandiari, N. Azarpira, H. Niknahad, Mitochondria pro-
Krebs cycle) enzymes and mitochondrial respiration complexes. On the tection as a mechanism underlying the hepatoprotective effects of glycine in
other hand, other vital organs and tissues (e.g., kidneys, skeletal muscle, cholestatic mice, Biomed. Pharmacother 97 (Supplement C) (2018) 1086–1095.
and heart) could be affected by conditions such as cholestasis/cirrhosis [23] A.P. Rolo, P.J. Oliveira, A.J.M. Moreno, C.M. Palmeira, Bile acids affect liver
mitochondrial bioenergetics: possible relevance for cholestasis therapy, Toxicol.
[106,107]. Hence, investigating the potential protective properties of Sci. 57 (1) (2000) 177–185.
the molecules such as betaine against these complications could be the [24] A. Arduini, G. Serviddio, A.M. Tormos, M. Monsalve, J. Sastre, Mitochondrial
subject of further research. dysfunction in cholestatic liver diseases, Front. Biosci. 4 (2012) 2233–2252.
[25] C.M. Palmeira, A.P. Rolo, Mitochondrially-mediated toxicity of bile acids,
Toxicology 203 (1–3) (2004) 1–15.
Declaration of interest [26] E. Kathirvel, K. Morgan, G. Nandgiri, B.C. Sandoval, M.A. Caudill, T. Bottiglieri,
S.W. French, T.R. Morgan, Betaine improves nonalcoholic fatty liver and asso-
ciated hepatic insulin resistance: a potential mechanism for hepatoprotection by
The authors declare no conflicts of interest. betaine, Am. J. Physiol. 299 (5) (2010) G1068–G1077.
[27] D. Graf, A.K. Kurz, R. Reinehr, R. Fischer, G. Kircheis, D. Häussinger, Prevention of
Acknowledgment bile acid-induced apoptosis by betaine in rat liver, Hepatology 36 (4 Pt. 1)) (2002)
829–839.
[28] Q. Garrett, N. Khandekar, S. Shih, J.L. Flanagan, P. Simmons, J. Vehige,
The authors thank Pharmaceutical Sciences Research Center of M.D.P. Willcox, Betaine stabilizes cell volume and protects against apoptosis in
Shiraz University of Medical Sciences for providing technical and fi- human corneal epithelial cells under hyperosmotic stress, Exp. Eye Res. 108
(Supplement C) (2013) 33–41.
nancial support of the current investigation (Grant number: 12046/
[29] L. Moezi, R. Heidari, Z. Amirghofran, A.A. Nekooeian, A. Monabati, A.R. Dehpour,
10930). Enhanced anti-ulcer effect of pioglitazone on gastric ulcers in cirrhotic rats: the
role of nitric oxide and IL-1b, Pharmacol. Rep. 65 (134) (2013) 134–143.
References [30] M.J. Tuñón, M. Alvarez, J.M. Culebras, J. González-Gallego, An overview of an-
imal models for investigating the pathogenesis and therapeutic strategies in acute
hepatic failure, W. J. Gastroenterol. 15 (25) (2009).
[1] S.A.S. Craig, Betaine in human nutrition, Am. J. Clin. Nut. 80 (3) (2004) 539–549. [31] S. Ali, K.A. Ansari, M.A. Jafry, H. Kabeer, G. Diwakar, Nardostachys jatamansi
[2] V. Preedy, Betaine: Chemistry, Analysis, Function and Effects, Royal Society protects against liver damage induced by thioacetamide in rats, J.
Chemistry, 2015. Ethnopharmacol. 71 (3) (2000) 359–363.
[3] M. Ahn, J.S. Park, S. Chae, S. Kim, C. Moon, J.W. Hyun, T. Shin, Hepatoprotective [32] R. Bruck, H. Aeed, H. Shirin, Z. Matas, L. Zaidel, Y. Avni, Z. Halpern, The hydroxyl
effects of Lycium chinense Miller fruit and its constituent betaine in CCl4-induced radical scavengers dimethylsulfoxide and dimethylthiourea protect rats against
hepatic damage in rats, Acta Histochem. 116 (6) (2014) 1104–1112. thioacetamide-induced fulminant hepatic failure, J. Hepatol. 31 (1) (1999) 27–38.
[4] J. Balkan, F.H. Parldar, S. Dogru-Abbasoglu, G. Aykaç-Toker, M. Uysal, The effect [33] A. Jamshidzadeh, R. Heidari, S. Mohammadi-Samani, N. Azarpira, A. Najbi,
of taurine or betaine pretreatment on hepatotoxicity and prooxidant status in- P. Jahani, N. Abdoli, A comparison between the nephrotoxic profile of gentamicin
duced by lipopolysaccharide treatment in the liver of rats, Eur. J. Gastroenterol. and gentamicin nanoparticles in mice, J. Biochem. Mol. Toxicol. 29 (2) (2015)
Hepatol. 17 (9) (2005) 917. 57–62.
[5] Y.S. Jung, S.J. Kim, D.Y. Kwon, C.W. Ahn, Y.S. Kim, D.W. Choi, Y.C. Kim, [34] R. Gupta, D.K. Dubey, G.M. Kannan, S.J.S. Flora, Concomitant administration of
Alleviation of alcoholic liver injury by betaine involves an enhancement of anti- Moringa oleifera seed powder in the remediation of arsenic-induced oxidative
oxidant defense via regulation of sulfur amino acid metabolism, Food Chem. stress in mouse, Cell Biol. Int. 31 (1) (2007) 44–56.
Toxicol. 62 (2013) 292–298. [35] R. Heidari, N. Esmailie, N. Azarpira, A. Najibi, H. Niknahad, Effect of thiol-re-
[6] Z. Wang, T. Yao, M. Pini, Z. Zhou, G. Fantuzzi, Z. Song, Betaine improved adipose ducing agents and antioxidants on sulfasalazine-induced hepatic injury in nor-
tissue function in mice fed a high-fat diet: a mechanism for hepatoprotective effect motermic recirculating isolated perfused rat liver, Toxicol. Res. 32 (2) (2016)
of betaine in nonalcoholic fatty liver disease, Am. J. Physiol. 298 (5) (2010) 133–140.
G634–G642. [36] D.J. Socci, K.B. Bjugstad, H.C. Jones, J.V. Pattisapu, G.W. Arendash, Evidence that
[7] C. Ji, N. Kaplowitz, Betaine decreases hyperhomocysteinemia, endoplasmic re- oxidative stress is associated with the pathophysiology of inherited hydrocephalus
ticulum stress, and liver injury in alcohol-fed mice, Gastroenterology 124 (5) in the H-Tx rat model, Exp. Neurol. 155 (1) (1999) 109–117.
(2003) 1488–1499. [37] H. Niknahad, R. Heidari, R. Firuzi, F. Abazari, M. Ramezani, N. Azarpira,
[8] J. Oliva, F. Bardag-Gorce, B. Tillman, S.W. French, Protective effect of quercetin, M. Hosseinzadeh, A. Najibi, A. Saeedi, Concurrent inflammation augments anti-
EGCG, catechin and betaine against oxidative stress induced by ethanol in vitro, malarial drugs-induced liver injury in rats, Ad. Pharm. Bull. 6 (4) (2016) 617–625.
Exp. Mol. Pathol. 90 (3) (2011) 295–299. [38] R. Heidari, A. Jamshidzadeh, H. Niknahad, F. Safari, H. Azizi, N. Abdoli,
[9] B.L. Copple, H. Jaeschke, C.D. Klaassen, Oxidative stress and the pathogenesis of M.M. Ommati, F. Khodaei, A. Saeedi, A. Najibi, The hepatoprotection provided by
cholestasis, Semin. Liver Dis. 30 (2) (2010) 195–204. taurine and Glycine against antineoplastic drugs induced liver injury in an ex vivo
[10] H. Jaeschke, G.J. Gores, A.I. Cederbaum, J.A. Hinson, D. Pessayre, J.J. Lemasters, model of normothermic recirculating isolated perfused rat liver, Trend. Pharm.
Mechanisms of hepatotoxicity, Toxicol. Sci. 65 (2) (2002) 166–176. Sci. 2 (1) (2016) 59–76.
[11] H. Jaeschke, M.R. McGill, A. Ramachandran, Oxidant stress, mitochondria, and [39] R. Heidari, M. Rasti, B. Shirazi Yeganeh, H. Niknahad, A. Saeedi, A. Najibi,
cell death mechanisms in drug-induced liver injury: lessons learned from acet- Sulfasalazine-induced renal and hepatic injury in rats and the protective role of
aminophen hepatotoxicity, Drug Metab. Rev. 44 (1) (2012) 88–106. taurine, BioImpacts 6 (1) (2016) 3–8.
[12] E. Mormone, J. George, N. Nieto, Molecular pathogenesis of hepatic fibrosis and [40] V. Katalinic, D. Modun, I. Music, M. Boban, Gender differences in antioxidant
current therapeutic approaches, Chem.-Biol. Interact. 193 (3) (2011) 225–231. capacity of rat tissues determined by 2,2’-azinobis (3-ethylbenzothiazoline 6-sul-
[13] C. Stephens, R.J. Andrade, M.I. Lucena, Mechanisms of drug-induced liver injury, fonate; ABTS) and ferric reducing antioxidant power (FRAP) assays, Comp.
Curr. Opin. Allergy Clin. Immunol. 14 (4) (2014) 286–292. Biochem. Physiol. 140 (1) (2005) 47–52.
[14] M.R. Duchen, Mitochondria in health and disease: perspectives on a new mi- [41] M. Alía, C. Horcajo, L. Bravo, L. Goya, Effect of grape antioxidant dietary fiber on
tochondrial biology, Mol. Asp. Med. 25 (4) (2004) 365–451. the total antioxidant capacity and the activity of liver antioxidant enzymes in rats,
[15] S.W.G. Tait, D.R. Green, Mitochondria and cell death: outer membrane permea- Nutr. Res. 23 (9) (2003) 1251–1267.
bilization and beyond, Nat. Rev. Mol. Cell Biol. 11 (9) (2010) 621–632. [42] R. Heidari, L. Moezi, B. Asadi, M.M. Ommati, N. Azarpira, Hepatoprotective effect
[16] G. Labbe, D. Pessayre, B. Fromenty, Drug-induced liver injury through mi- of boldine in a bile duct ligated rat model of cholestasis/cirrhosis,
tochondrial dysfunction: mechanisms and detection during preclinical safety stu- PharmaNutrition 5 (3) (2017) 109–117.
dies, Fundam. Clin. Pharmacol. 22 (4) (2008) 335–353. [43] A.A. Caro, L.W. Adlong, S.J. Crocker, M.W. Gardner, E.F. Luikart, L.U. Gron, Effect
[17] R. Heidari, H. Niknahad, A. Jamshidzadeh, M.A. Eghbal, N. Abdoli, An overview of garlic-derived organosulfur compounds on mitochondrial function and integrity
on the proposed mechanisms of antithyroid drugs-induced liver injury, Adv. in isolated mouse liver mitochondria, Toxicol. Lett. 214 (2) (2012) 166–174.

84
R. Heidari et al. Biomedicine & Pharmacotherapy 103 (2018) 75–86

[44] P. Zhao, T.F. Kalhorn, J.T. Slattery, Selective mitochondrial glutathione depletion Abbasoğlu, M. Uysal, High-fat diet plus carbon tetrachloride-induced liver fibrosis
by ethanol enhances acetaminophen toxicity in rat liver, Hepatology 36 (2) (2002) is alleviated by betaine treatment in rats, Int. Immunopharmacol. 39 (2016)
326–335. 199–207.
[45] T. Mosmann, Rapid colorimetric assay for cellular growth and survival: applica- [73] R. Deminice, R.Pd. Silva, S.G. Lamarre, K.B. Kelly, R.L. Jacobs, M.E. Brosnan,
tion to proliferation and cytotoxicity assays, J. Immunol. Methods 65 (1) (1983) J.T. Brosnan, Betaine supplementation prevents fatty liver induced by a high-fat
55–63. diet: effects on one-carbon metabolism, Amino Acids 47 (4) (2015) 839–846.
[46] R. Heidari, F. Jafari, F. Khodaei, B. Shirazi Yeganeh, H. Niknahad, The mechanism [74] M.F. Abdelmalek, P. Angulo, R.A. Jorgensen, P.B. Sylvestre, K.D. Lindor, Betaine, a
of valproic acid-induced Fanconi syndrome involves mitochondrial dysfunction promising new agent for patients with nonalcoholic steatohepatitis: results of a
and oxidative stress in rat kidney, Nephrology (Carlton, Vic.) 23 (4) (2018) pilot study, Am. J. Gastroenterol. 96 (9) (2001) 2711–2717.
351–361. [75] H. Hagar, W. Al Malki, Betaine supplementation protects against renal injury in-
[47] R. Heidari, V. Taheri, H.R. Rahimi, B.S. Yeganeh, H. Niknahad, A. Najibi, duced by cadmium intoxication in rats: role of oxidative stress and caspase-3,
Sulfasalazine-induced renal injury in rats and the protective role of thiol-re- Environ. Toxicol. Pharmacol. 37 (2) (2014) 803–811.
ductants, Ren. Fail. 38 (1) (2016) 137–141. [76] H. Hagar, A.E. Medany, R. Salam, G.E. Medany, O.A. Nayal, Betaine supple-
[48] A. Jamshidzadeh, H. Niknahad, R. Heidari, M. Zarei, M.M. Ommati, F. Khodaei, mentation mitigates cisplatin-induced nephrotoxicity by abrogation of oxidative/
Carnosine protects brain mitochondria under hyperammonemic conditions: re- nitrosative stress and suppression of inflammation and apoptosis in rats, Exp.
levance to hepatic encephalopathy treatment, PharmaNutrition 5 (2) (2017) Toxicol. Pathol. 67 (2) (2015) 133–141.
58–63. [77] F. Erman, J. Balkan, U. Çevikbaş, N. Koçak-Toker, M. Uysal, Betaine or taurine
[49] H. Niknahad, A. Jamshidzadeh, R. Heidari, Z. Hosseini, K. Mobini, F. Khodaei, administration prevents fibrosis and lipid peroxidation induced by rat liver by
M.M. Ommati, N. Abdoli, N. Keshavarz, M. Bazyari, A. Najibi, Paradoxical effect of ethanol plus carbon tetrachloride intoxication, Amino Acids 27 (2) (2004)
methimazole on liver mitochondria: in vitro and in vivo, Toxicol. Lett. 259 (2016) 199–205.
108–115. [78] B. Ganesan, S. Buddhan, R. Anandan, R. Sivakumar, R. AnbinEzhilan, Antioxidant
[50] H. Niknahad, A. Jamshidzadeh, R. Heidari, M. Zarei, M.M. Ommati, Ammonia- defense of betaine against isoprenaline-induced myocardial infarction in rats, Mol.
induced mitochondrial dysfunction and energy metabolism disturbances in iso- Biol. Rep. 37 (3) (2009) 1319–1327.
lated brain and liver mitochondria, and the effect of taurine administration: re- [79] S.K. Kim, J.M. Seo, Y.R. Chae, Y.S. Jung, J.H. Park, Y.C. Kim, Alleviation of di-
levance to hepatic encephalopathy treatment, Clin. Exp. Hepatol. 3 (3) (2017) methylnitrosamine-induced liver injury and fibrosis by betaine supplementation in
141–151. rats, Chem.-Biol. Interact. 177 (3) (2009) 204–211.
[51] M.M. Ommati, R. Heidari, A. Jamshidzadeh, M.J. Zamiri, Z. Sun, S. Sabouri, [80] S. Cayley, B.A. Lewis, M.T. Record, Origins of the osmoprotective properties of
J. Wang, F. Ahmadi, N. Javanmard, K. Seifi, S. Mousapour, B.S. Yeganeh, Dual betaine and proline in Escherichia coli K-12, J. Bacteriol. 174 (5) (1992)
effects of sulfasalazine on rat sperm characteristics, spermatogenesis, and ster- 1586–1595.
oidogenesis in two experimental models, Toxicol. Lett. 284 (2018) 46–55. [81] M. Wettstein, D. Haussinger, Cytoprotection by the osmolytes betaine and taurine
[52] M.M. Ommati, N. Tanideh, B. Rezakhaniha, J. Wang, S. Sabouri, M. Vahedi, in ischemia- reoxygenation injury in the perfused rat liver, Hepatology 26 (6)
B. Dormanesh, O. Koohi Hosseinabadi, F. Rahmanifar, S. Moosapour, A. Akhlaghi, (1997) 1560–1566.
R. Heidari, M.J. Zamiri, Is immunosuppression, induced by neonatal thymectomy, [82] A.J. Barak, H.C. Beckenhauer, M. Junnila, D.J. Tuma, Dietary betaine promotes
compatible with poor reproductive performance in adult male rats? Andrology 6 generation of hepatic S-adenosylmethionine and protects the liver from ethanol-
(1) (2018) 199–213. induced fatty infiltration, Alcoholism: Clin. Exp. Res. 17 (3) (1993) 552–555.
[53] M.M. Bradford, A rapid and sensitive method for the quantitation of microgram [83] S.K. Kim, Y.C. Kim, Effects of betaine supplementation on hepatic metabolism of
quantities of protein utilizing the principle of protein-dye binding, Analyt. sulfur-containing amino acids in mice, J. Hepatol. 42 (6) (2005) 907–913.
Biochem. 72 (1) (1976) 248–254. [84] J.M. Mato, F.J. Corrales, S.C. Lu, M.A. Avila, S-Adenosylmethionine: a control
[54] M. Pinzani, K. Rombouts, Liver fibrosis: from the bench to clinical targets, Dig. switch that regulates liver function, FASEB J. 16 (1) (2002) 15–26.
Liver Dis. 36 (4) (2004) 231–242. [85] Q.M. Anstee, C.P. Day, S-Adenosylmethionine (SAMe) therapy in liver disease: a
[55] C. Ara, H. Kirimlioglu, A.B. Karabulut, S. Coban, S. Ay, M. Harputluoglu, review of current evidence and clinical utility, J. Hepatol. 57 (5) (2012)
V. Kirimlioglu, S. Yilmaz, Protective effect of resveratrol against oxidative stress in 1097–1109.
cholestasis, J. Surg. Res. 127 (2) (2005) 112–117. [86] R.L. Jacobs, J.N. Veen, D.E. Vance, Finding the balance: the role of S-adeno-
[56] H. Shapiro, M. Ashkenazi, N. Weizman, M. Shahmurov, H. Aeed, R. Bruck, sylmethionine and phosphatidylcholine metabolism in development of nonalco-
Curcumin ameliorates acute thioacetamide-induced hepatotoxicity, J. holic fatty liver disease, Hepatology 58 (4) (2013) 1207–1209.
Gastroenterol. Hepatol. 21 (2) (2006) 358–366. [87] A.A. Caro, A.I. Cederbaum, Antioxidant properties of S-adenosyl-l-methionine in
[57] C.G. Tag, S. Sauer-Lehnen, S. Weiskirchen, E. Borkham-Kamphorst, R.H. Tolba, Fe2+-initiated oxidations, Free Radic. Biol. Med. 36 (10) (2004) 1303–1316.
F. Tacke, R. Weiskirchen, Bile duct ligation in mice: induction of inflammatory [88] M. Rizzardini, M. Carelli, M.R. Cabello Porras, L. Cantoni, Mechanisms of en-
liver injury and fibrosis by obstructive cholestasis, J. Vis. Exp. (2015) 96. dotoxin-induced haem oxygenase mRNA accumulation in mouse liver: synergism
[58] E. Krones, M. Wagner, K. Eller, A.R. Rosenkranz, M. Trauner, P. Fickert, Bile acid- by glutathione depletion and protection by N-acetylcysteine, Biochem. J. 304 (Pt
induced cholemic nephropathy, Dig. Dis. 33 (3) (2015) 367–375. 2) (1994) 477–483.
[59] T. Li, J.Y.L. Chiang, Bile acid-induced liver injury in cholestasis, Cellular Injury in [89] Z. Ming, Y.-j. Fan, X. Yang, W.W. Lautt, Synergistic protection by S-adeno-
Liver Diseases, Springer, Cham, 2017, pp. 143–172. sylmethionine with vitamins C and E on liver injury induced by thioacetamide in
[60] R.H. Palmer, Bile acids, liver injury, and liver disease, Arch. Intern. Med. 130 (4) rats, Free Radic. Biol. Med. 40 (4) (2006) 617–624.
(1972) 606–617. [90] M.-T. Tsai, C.-Y. Chen, Y.-H. Pan, S.-H. Wang, H.J. Mersmann, S.-T. Ding,
[61] R.J. Sokol, M. Devereaux, R. Dahl, E. Gumpricht, “Let there be bile”–under- Alleviation of carbon-tetrachloride-induced liver injury and fibrosis by betaine
standing hepatic injury in cholestasis, J. Pediatr. Gastroenterol. Nutr. 43 (Suppl. 1) supplementation in chickens, Evid. Based Complement. Alternat. Med. (2015).
(2006) S4–S9. [91] R.A. Roth, J.P. Luyendyk, J.F. Maddox, P.E. Ganey, Inflammation and drug idio-
[62] A.P. Rolo, C.M. Palmeira, K.B. Wallace, Mitochondrially mediated synergistic cell syncrasy—is there a connection? J. Pharmacol. Exp. Ther. 307 (1) (2003) 1–8.
killing by bile acids, Biochim. Biophys. Acta 1637 (1) (2003) 127–132. [92] W. Zou, R.A. Roth, P.E. Ganey, A.V. Lyubimov, Animal models of idiosyncratic,
[63] A. Jamshidzadeh, R. Heidari, Z. Latifpour, M.M. Ommati, N. Abdoli, S. Mousavi, drug-induced liver injury: emphasis on the inflammatory stress hypothesis,
N. Azarpira, A. Zarei, M. Zarei, B. Asadi, M. Abasvali, Y. Yeganeh, F. Jafari, Encyclopedia of Drug Metabolism and Interactions, John Wiley & Sons, Inc., 2011.
A. Saeedi, A. Najibi, E. Mardani, Carnosine ameliorates liver fibrosis and hyper- [93] D.H. Kim, B. Sung, Y.J. Kang, J.Y. Jang, S.Y. Hwang, Y. Lee, M. Kim, E. Im, J.-
ammonemia in cirrhotic rats, Clin. Res. Hepatol. Gastroenterol. 41 (4) (2017) H. Yoon, C.M. Kim, H.Y. Chung, N.D. Kim, Anti-inflammatory effects of betaine on
424–434. AOM/DSS‑induced colon tumorigenesis in ICR male mice, Int. J. Oncol. 45 (3)
[64] M. Jain, S. Rivera, E.A. Monclus, L. Synenki, A. Zirk, J. Eisenbart, C. Feghali- (2014) 1250–1256.
Bostwick, G.M. Mutlu, G.R.S. Budinger, N.S. Chandel, Mitochondrial reactive [94] E.K. Go, K.J. Jung, J.Y. Kim, B.P. Yu, H.Y. Chung, Betaine suppresses proin-
oxygen species regulate transforming growth factor-β signaling, J. Biol. Chem. 288 flammatory signaling during aging: the involvement of nuclear factor-κB via nu-
(2) (2013) 770–777. clear factor-inducing kinase/IκB kinase and mitogen-activated protein kinases, J.
[65] M.J. Parsons, D.R. Green, Mitochondria in cell death, Essays Biochem. 47 (2010) Gerontol. 60 (10) (2005) 1252–1264.
99–114. [95] R.R. Watson, Foods and Dietary Supplements in the Prevention and Treatment of
[66] A.P. Halestrap, E. Doran, J.P. Gillespie, A. O’Toole, Mitochondria and cell death, Disease in Older Adults, Academic Press, 2015.
Biochem. Soc. Trans. 28 (1) (2000) 11. [96] R.K. Moreira, Hepatic stellate cells and liver fibrosis, Arch. Pathol. Lab. Med. 131
[67] P. Bernardi, L. Scorrano, R. Colonna, V. Petronilli, F. Di Lisa, Mitochondria and (11) (2007) 1728–1734.
cell death, Eur. J. Biochem. 264 (3) (1999) 687–701. [97] R. Safadi, S.L. Friedman, Hepatic fibrosis–role of hepatic stellate cell activation,
[68] M.R. Duchen, Mitochondria and calcium: from cell signalling to cell death, J. Med. Gen. Med. 4 (3) (2002) 27.
Physiol. 529 (1) (2000) 57–68. [98] M. Junnila, T. Rahko, A. Sukura, L.A. Lindberg, Reduction of carbon tetrachloride-
[69] N. Abdoli, R. Heidari, Y. Azarmi, M.A. Eghbal, Mechanisms of the statins cyto- induced hepatotoxic effects by oral administration of betaine in male Han-Wistar
toxicity in freshly isolated rat hepatocytes, J. Biochem. Mol. Toxicol. 27 (6) (2013) rats: a morphometric histological study, Vet. Pathol. 37 (3) (2000) 231–238.
287–294. [99] I. Lee, Betaine is a positive regulator of mitochondrial respiration, Biochem.
[70] R. Heidari, H. Niknahad, A. Jamshidzadeh, N. Abdoli, Factors affecting drug-in- Biophys. Res. Commun. 456 (2) (2015) 621–625.
duced liver injury: antithyroid drugs as instances, Clin. Mol. Hepatol. 20 (3) [100] B. Ganesan, R. Rajesh, R. Anandan, N. Dhandapani, Biochemical studies on the
(2014) 237–248. protective effect of betaine on mitochondrial function in experimentally induced
[71] S.A. Kempson, Y. Zhou, N.C. Danbolt, The betaine/GABA transporter and betaine: myocardial infarction in rats, J. Health Sci. 53 (6) (2007) 671–681.
roles in brain, kidney, and liver, Front. Physiol. 5 (2014). [101] A.R. Im, Y.-H. Kim, M.R. Uddin, S. Chae, H.W. Lee, Y.H. Kim, Y.S. Kim, M.-Y. Lee,
[72] İ. Bingül, A.F. Aydın, C. Başaran-Küçükgergin, I. Doğan-Ekici, J. Çoban, S. Doğru- Betaine protects against rotenone-induced neurotoxicity in PC12 cells, Cell. Mol.

85
R. Heidari et al. Biomedicine & Pharmacotherapy 103 (2018) 75–86

Neurobiol. 33 (5) (2013) 625–635. mitochondrial permeability transition, Hepatology 33 (3) (2001) 616–626.
[102] K.K. Kharbanda, S.L. Todero, A.L. King, N.A. Osna, B.L. McVicker, D.J. Tuma, [105] P.S. Brookes, Y. Yoon, J.L. Robotham, M.W. Anders, S.-S. Sheu, Calcium, ATP, and
J.L. Wisecarver, S.M. Bailey, Betaine treatment attenuates chronic ethanol-in- ROS: a mitochondrial love-hate triangle, Am. J. Physiol. 287 (4) (2004)
duced hepatic steatosis and alterations to the mitochondrial respiratory chain C817–C833.
proteome, Int. J. Hepatol. (2011) 2012. [106] M. Kalafateli, C. Konstantakis, K. Thomopoulos, C. Triantos, Impact of muscle
[103] D. Nash, L.G. Paleg, J.J. Wiskich, Effect of proline, betaine and some other solutes wasting on survival in patients with liver cirrhosis, W. J. Gastroenterol. 21 (24)
on the heat stability of mitochondrial enzymes, Funct. Plant Biol. 9 (1) (1982) (2015) 7357–7361.
47–57. [107] E. Krones, M. Wagner, K. Eller, A.R. Rosenkranz, M. Trauner, P. Fickert, Bile acid-
[104] B. Yerushalmi, R. Dahl, M.W. Devereaux, E. Gumpricht, R.J. Sokol, Bile acid‐- induced cholemic nephropathy, Dig. Dis. 33 (3) (2015) 367–375.
induced rat hepatocyte apoptosis is inhibited by antioxidants and blockers of the

86

You might also like