Open Access

Berberis vulgaris extract/β-cyclodextrin complex increases protection of hepatic cells via suppression of apoptosis and lipogenesis pathways

  • Authors:
    • Alexandra Ivan
    • Hildegard Herman
    • Cornel Balta
    • Daniel I. Hadaruga
    • Ciprian‑Valentin Mihali
    • Aurel Ardelean
    • Anca Hermenean
  • View Affiliations

  • Published online on: March 20, 2017     https://doi.org/10.3892/etm.2017.4240
  • Pages: 2143-2150
  • Copyright: © Ivan et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Berberis vulgaris (Bv) is well known worldwide for its healing properties. However, limited information is available concerning its mechanism of action and the increased hepatoprotective activity of formulated extracts. This study evaluated the protective effect of Bv bark extract against CCl4‑induced cytotoxicity in Huh7 cells, as well whether β-cyclodextrin complexation of the extract resulted in increased hepatoprotective effects. Huh7 cells were incubated for 48 h with 5, 7.5 and 10 µg/ml of unformulated or formulated Bv extract alone and in co‑treatment with CCl4. The effects on Huh7 cell growth and apoptosis were evaluated by MTT assay, caspase‑3/7 activity and caspase-3 expression, whereas fatty acid changes were investigated by Oil red O staining and the detection of peroxisome proliferator-activated receptor-γ (PPARγ) expression using immunofluorescence. Ultrastructural alterations were observed by electron microscopy. The MTT assay showed that co‑exposure to CCl4 and 7.5 µg/ml formulated extract led to a 1.25‑fold increase in cell viability compared with the non‑formulated extract. Caspase‑3/7 activity decreased by 50% and 70% following co-treatment with unformulated or formulated extract, compared with that in cells treated with CCl4 alone. Furthermore, hepatocyte ultrastructure was protected from CCl4‑induced injury in the two co‑treated groups, intracytoplasmic lipid accumulation decreased significantly and PPARγ expression was restored, in comparison with CCl4-treated cells alone. Formulated and unformulated extracts were efficient against the anti-proliferative and pro‑apoptotic actions of CCl4 through suppression of CCl4‑induced caspase‑3 activation and lipid accumulation. The protective effect of the formulated extract was more pronounced than that of the unformulated one, which may be due to its increased solubility.

Introduction

Carbon tetrachloride (CCl4) is an experimental hepatotoxicant, able to induce acute liver injury characterized by necrosis and steatosis (1,2). Once the metabolism of CCl4 is initiated, its toxicity is determined by chain chemical reactions that take place in the body. The initial step reaction is a reduction and heterolytic cleavage of CCl4 to form a trichloromethyl radical (•CCl3) (3). This radical reacts either directly with cellular macromolecules or with oxygen; reaction with the latter forms the trichloromethylperoxyl radical (•OOCCl3), which acts more rapidly on lipids than does •CCl3 (4). Production of excessive amounts of reactive oxygen species (ROS) generates oxidative stress which induces major cellular perturbations, such as alterations in the structures of protein and nucleic acids, increases in intracellular free calcium levels, perturbation of membrane permeability, lipid peroxidation and finally cell death (5). In order to minimize the effects of ROS, cells have an antioxidant defense system which includes non-enzymatic antioxidants and enzymes such as superoxide dismutase, catalase and glutathione peroxidase (6). CCl4 administration destabilizes this defense system and decreases superoxide dismutase, catalase and glutathione peroxidase activity (7).

Berberis vulgaris L. (Berberidaceae) (Bv) has been well known worldwide for its healing properties for >2,500 years (8). The bioactive components are represented by several alkaloid constituents, such as berberine, berbamine and palmatine, which confer healing properties to Berberis extracts (9). Berberine is the most important isoquinoline alkaloid, obtained mostly from the roots and bark of Berberis sp. (10). Berberine is known for its multiple pharmacological properties, such as antimicrobial (11), antitumor (12) and anti-inflammatory effects (13,14). Berberine is also known for its dose-dependent hepatoprotective effects on CCl4-induced liver damage, due to its antioxidant effects (15).

This study was carried out to evaluate for the first time the increased protective effect of a formulation of Bv bark extract in β-cyclodextrin (β-CD) against CCl4-induced cytotoxicity in Huh7 cells. This natural complex was designed for use in oral formulations, in order to increase the solubility, dissolution, bioavailability, safety and stability of the extract via certain properties of β-CD, including its resistance to hydrolysis by human salivary and pancreatic amylases (16).

Materials and methods

Complex of Bv bark extract and β-CD

Samples of Bv were collected from the Botanical Garden of Vasile Goldis Western University of Arad (Arad, Romania) during October 2008, and certified at the herbarium within the Faculty of Natural Sciences, where a voucher specimen already exists. The synthesis and characterization of the Bv bark extract/β-cyclodextrin complex used in this study have been previously reported by the authors (2).

Cell culture

The study was carried out using as model a Huh7 human hepatoma cell line (American Type Culture Collection, Manassas, VA, USA). The Huh7 cell line was chosen due to its metabolic similarities to normal hepatocytes and in order to avoid the variability and short life spans of primary human hepatocytes (17). Cells were grown in Dulbecco's modified Eagle's medium (DMEM; Sigma-Aldrich, Irvine, UK) supplemented with 10% fetal calf serum (FCS; Sigma-Aldrich, Steinheim, Germany), and 1% penicillin-streptomycin (Pen/Strep, 10,000 IU/ml; PromoCell GmbH, Heidelberg, Germany) in a humidified atmosphere with 5% CO2, at 37°C.

Cell treatment

Cells were plated at a density of 104 cells/cm2 with DMEM medium (high glucose, supplemented with 10% FCS) and allowed to attach overnight at 37°C. The CCl4 concentration (0.1 mM) used for cell culture co-treatment was previously determined and chosen due to its ability to induce up to 75% cell culture mortality. Three concentrations (5, 7.5 and 10 µg/ml) of unformulated and nanoencapsulated β-CD Bv bark extract were tested for protection against CCl4-induced cytotoxicity in the Huh7 cell line. Each experiment was performed in triplicate under 48 h of exposure. Stock solutions were prepared fresh to avoid oxidation. The Bv extracts were dissolved in dimethylsulfoxide (DMSO) and diluted with DMEM to the desired concentrations prior to use, while DMSO alone was used as vehicle control.

3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) cytotoxicity assays

The MTT assay was used to detect the cytotoxicity of unformulated and formulated Bv extract. Cells were seeded into 96-well plates and allowed to attach overnight. A series of unformulated and formulated extracts were added (5, 7.5 and 10 µg/ml), alone or together with 0.1 mM CCl4, followed by 48 h incubation. All experiments were conducted in parallel with a control. The MTT assay was performed using a commercially available MTT assay (MTT base; Sigma-Aldrich, St. Louis, MO, USA) according to the manufacturer's protocol. The absorbance (abs) was measured at 565 nm, using a Tecan Infinite F200 microplate reader (Tecan, Männedorf, Switzerland). The cell survival rate was calculated as follows: Survival rate (%) = (Abs treatment - abs blank)/(Abs control - Abs blank) × 100.

Caspase-3 and −7 activities

Total caspase-3 and −7 activities were measured using an Apo-ONE Homogeneous Caspase-3/7 assay kit (Promega Corporation, Madison, WI, USA). Following the various treatments, 100 µl Apo-ONE Caspase-3/7 reagent (substrate and buffer in the ratio of 1:100) were added to each well of a 96-well plate. After 1 h incubation in the dark at room temperature, the fluorescence of each well was measured at 485–520 nm (Fluoroskan Ascent FL; Thermo Fisher Scientific, Inc., Waltham, MA, USA).

Immunofluorescence

Expression levels of caspase-3 and peroxisome proliferator-activated receptor-γ (PPARγ) were assessed by immunofluorescence using a rabbit polyclonal caspase-3 antibody (orb10273; Biorbyt Ltd., Cambridge, UK) 1:200 dilution, or PPARγ antibody (sc-7196; Santa Cruz Biotechnology, Inc., Dallas, TX, USA) 1:50 dilution and AlexaFluor 488-labeled chicken anti-rabbit IgG secondary antibody (A-21441; Invitrogen; Thermo Fisher Scientific, Inc.). Cells were treated with unformulated and nanoencapsulated β-CD extract for 48 h prior to fixing with cold 100% methanol at −20°C, for 10 min. The cells treated under experimental conditions were fixed with cold 100% methanol at −20°C for 10 min. Blocking of non-specific binding was conducted by incubation with 1% bovine serum albumin (Antibodies Online Inc., Atlanta, GA, USA) and 0.1% Triton X (Sigma-Aldrich; Merck Millipore) for 1 h at room temperature. Following blocking, cells were incubated overnight at 4°C with corresponding dilutions of the primary antibodies. Following overnight incubation, the cells were washed in PBS and incubated with secondary antibody for 1 h at room temperature. Nucleus counterstaining was performed with 10 µg/ml Hoechst 33285 working solution (861405; Sigma-Aldrich, Buchs, Switzerland) for 60 sec, followed by analysis using a Leica TCS SP8 confocal microscope (Leica Microsystems GmbH, Wetzlar, Germany). The fluorescence intensity of PPARγ- and caspase 3-positive hepatocytes was quantified by automated counting performed using an image analysis system (ImageJ; National Institutes of Health, Bethesda, MD, USA). Three fields were selected randomly from each section, and a total of 100 hepatocytes were evaluated in every field.

Oil Red O staining

Intracellular accumulation of lipid droplets was observed using Oil Red O staining. Cells were plated on Lab-Tek Chamber Slides (Nalge Nunc International, Penfield, NY, USA) for 24 h. After 24 h of seeding, cells were treated with unformulated and β-CD nanoencapsulated Bv extract, respectively, for 48 h. For staining, the cells were fixed with 4% formaldehyde for 10 min at 4°C. The fixed cultures were incubated with Oil Red O working solution for 20 min at room temperature. Oil Red O was provided in a staining kit (04–220923; Bio Optica, Milan, Italy). Nucleus counterstaining was performed with Mayer's hematoxylin provided in the same kit (04–220923; Bio Optica). After washing with tap water, the Permanox slides were mounted. Analysis was performed using an Olympus BX53 microscope (Olympus Corporation, Tokyo, Japan).

Electron microscopy

For electron microscopy, 1×106 cells were treated with unformulated and β-CD nanoencapsulated Bv extracts for 48 h. Following the drug treatments, cells were collected by trypsinization, followed by centrifugation at 240 × g. The obtained pellet was prefixed in 2.7% glutaraldehyde solution in 0.1 M phosphate buffer for 1.5 h, at 4°C, then washed in 0.15 M phosphate buffer (pH 7.2) and post-fixed in 2% osmic acid solution in 0.15 M phosphate buffer for 1 h at 4°C. Dehydration was performed in acetone, followed by inclusion in the epoxy embedding resin Epon 812. Blocks 70-nm thick were cut using an LKB ultramicrotome. The sections were doubly contrasted with solutions of uranyl acetate and lead citrate and analyzed under a Tecnai 12 Biotwin transmission electron microscope (FEI, Hillsboro, OR, USA).

Statistical analysis

Statistical analysis of the differences between the means of various treatment groups was performed using one-way analysis of variance. STATA statistical software (version 13.0; StataCorp LP, College Station, TX, USA) was used. P<0.05 was considered to indicate a statistically significant difference.

Results

Effects of unformulated and β-CD nanoencapsulated Bv extract on CCl4-induced cytotoxicity in Huh7 cells

To analyze the protective advantage of β-CD nanoencapsulated Bv extract against CCl4-induced cytotoxicity, cell viability in the Huh7 cell line was investigated. The cells were incubated with equivalent doses of formulated and unformulated Bv extract (5, 7.5 and 10 µg/ml Bv extract) for 48 h. As shown in Fig. 1A, the unformulated extract presented a dose-dependent significant cytotoxicity (P<0.001), whereas the formulated one did not. Due to the fact that the dose of 7.5 µg/ml extract presented moderate toxicity, it was chosen for the assessment of antioxidant activity. Following co-exposure of the cells to CCl4 and 7.5 µg/ml Bv nanoencapsulated extract, the cell viability was 1.25 fold higher compared with that of cells co-exposed to CCl4 and non-formulated Bv extract (Fig. 1B).

Effcts of unformulated and β-CD nanoencapsulated Bv extract on CCl4.-induced Huh7 cell apoptosis

In order to investigate the anti-apoptotic effects of Bv extracts, the caspase-3/7 activity (Fig. 2) as well as caspase-3 expression in control and experimental variants (Fig. 3) were evaluated. The caspase-3/7 activity increased significantly (P<0.05) by ~80% in the CCl4-treated cells compared with the control, whereas co-treatment with unformulated and formulated BV extract decreased it by 50 and 70%, respectively.

Analysis of confocal microscopy images showed that caspase-3 immunostaining was significantly increased in CCl4-treated cells compared with control cells. Under co-exposure to CCl4 with both formulated and unformulated Bv extract, signals were significantly reduced (P<0.001) compared with those in the CCl4-treated cells, and were reduced most strongly with the formulated Bv extract (P<0.001; Fig. 3).

Effects of unformulated and β-CD nanoencapsulated Bv extracts on CCl4.-induced fatty accumulation in Huh7 cells

Light microscopic evaluation of Huh7 cells stained with Oil Red O showed normal morphology of Huh7 cells and few accumulated intracellular lipid droplets in the control (Fig. 4A). Intracytoplasmic lipids increased significantly in the CCl4-treated cells without Bv extract, compared with the control (Fig. 4B). After 48 h of co-treatment with CCl4 and either Bv extract, HuH7 cells accumulated fewer lipid droplets compared with cells treated with CCl4 alone (Fig. 4C and D). In addition, the level of PPARγ expression decreased significantly (P<0.01) in CCl4-treated cells (Fig. 5), compared with control cells, and this reduction was attenuated in the cells co-treated with unformulated or formulated Bv extracts (Fig. 5).

Effects of unformulated and β-CD nanoencapsulated Bv extract on CCl4.-induced ultrastructural injuries of Huh7 cells

The ultrastructure of Huh7 cells was normal in the control group. Control cells showed integrated nuclear membranes, relatively homogeneous chromatin, and organelles with a normal appearance (Fig. 6A). Following treatment with 0.1 mM CCl4 for 48 h, Huh7 cells were characterized by irregular nuclear shapes, chromatin condensation into dense granules or blocks, vacuolar cytoplasm, and lipid droplet accumulation, together with altered mitochondria and rarefied cristae (Fig. 6B). Co-treatment with unformulated and formulated Bv extracts protected the hepatocyte ultrastructure from CCl4-induced injury; in the two BV groups the appearance of the Huh7 cells was similar to that of the control cells (Fig. 6C and D).

Discussion

Numerous herbs are used in traditional medicine to prevent and treat liver diseases as a result of hepatotoxicant action. Barberry root, bark, leaves and fruit have been used in traditional medicine to treat hepatic disorders, and berberine is generally thought to be responsible for their beneficial effects (18). Berberine itself is considered a potent antioxidant with a wide spectrum of applications (19); the biological activities and levels of safety should be established for the whole Bv extract and its formulations. Another important compound in barberry is berbamine, a bis-benzylisoquinoline alkaloid, which inhibits chemically-induced carcinogenesis (20).

To demonstrate the increased protective potential of β-CD nanoencapsulated Bv extract compared with unformulated Bv extract against CCl4-induced cytotoxicity, an MTT assay was performed in the present study. The notable cell mortality observed with formulated and unformulated extracts could be attributable to their content of berbamine, which inhibits Ca2+/calmodulin-dependent protein kinase II in liver cancer cells (21). CCl4 caused a reduction in cell viability of 62.43% compared with control. It is likely that a 7.5 µg/ml dose of unformulated or formulated extract presented a sufficient quantity of berberine to act as an antioxidant. The β-CD nanoencapsulated Bv extract displayed 1.25-fold higher protective effects compared with the unformulated form, probably due to its increased solubility. Previously, it has been demonstrated that the solubility of berberine hydrochloride was increased 5.27-fold by cyclodextrin complexation (22). In addition, Ziolkowski et al (23) have shown depletion of mitochondrial cholesterol in the presence methyl-β-cyclodextrin, which was associated with impaired bioenergetics and increased resistance to calcium chloride-induced swelling and subsequently prevented changes associated with early stage apoptosis.

In the present study 7.5 µg/ml Bv extract/β-CD complex exhibited no cytotoxicity whereas Kiss et al (24) showed that significant cell toxicity was induced by 50 mM β-CD; this difference in cytotoxicity may be explained by the CD concentration used being 75-fold lower than that in the previous study.

Apoptosis has been shown to be important in CCl4-induced hepatotoxicity (25). In the present study, apoptosis was confirmed by the detection of caspase-3 activation and ultrastructural analysis. CCl4 induced activation of the apoptotic signaling pathway via caspase-3, as shown in Figs. 2 and 3, consistent with previous in vivo studies that have demonstrated a similar apoptotic mechanism for this toxic agent (26). In addition, cell death could be initiated by the ability of CCl4 to arrest Huh7 cells in the G0/G1 phase. Also, it could decrease the proportion of cells in the S phase, which is associated with inhibition of DNA synthesis (27). Co-treatment with Bv/β-cyclodextrin complex significantly increased (P<0.001) the percentage of viable cells, which suggests that it reversed the cell cycle by increasing the proportion of cells in the S phase and inhibited the apoptotic process by suppression of CCl4-induced caspase-3 activation.

ROS generated by CCl4 exposure can cause changes in the inner mitochondrial membrane and loss of the mitochondrial transmembrane potential (28). As a result, cytochrome c and other mitochondrial proteins are released into the cytosol (29), activating caspase-9 and subsequently caspase-3, which cleaves a set of vital proteins and promotes the apoptotic degradation phase including DNA damage and morphological changes (30).

Apoptosis was also investigated by electron microscopy in the present study. The ultrastructural changes that were observed indicated the apoptosis of Huh7 cells due to the presence of CCl4, results that are in agreement with other results on HepG2 hepatocarcinoma cells exposed to fatty acid ethyl esters (31) or sarsasapogenin (32). In the present study, both formulated and unformulated Bv extracts were able to prevent ultrastructural injuries induced by CCl4 exposure.

Lipid droplets represent intracellular compartments able to store neutral lipids as triglycerides and cholesteryl esters. Each lipid droplet is surrounded by a phospholipid monolayer (33). The accumulation of neutral lipids (mainly triglycerides and cholesterol esters) into hepatocyte cytoplasm is associated with hepatic steatosis. This abnormal accumulation of lipid droplets may result from the release of free fatty acids (FFAs) by adipocytes, dietary intake, diminished hepatic export of FFAs, increased de novo lipogenesis or impaired β-oxidation of FFAs (34). In the present study, a massive accumulation of lipid droplets in the cytoplasm was observed after 48 h of CCl4 toxic exposure. The high increase in lipid concentration within the CCl4-treated Huh7 cells can be attributed to a reduction in fatty acid activation and cholesterol catabolism (35). Toxic injuries due to poisoning with compounds like CCl4 are expressed by fatty infiltration of the liver due to changes in low-density lipid synthesis (3,5,7). The use of unformulated and formulated Bv extracts exhibited a very good protective effect against the formation of lipid droplets due to CCl4 exposure, with slightly increased protective effects in the case of formulated Bv extracts.

PPARγ plays an important role as a regulator of lipid metabolism and is a key mediator of lipid storage and inflammation (36). The results of the current study present a number of intriguing observations. Firstly, in the CCl4-treated group intracellular lipid accumulation was observed, whereas a reduction in PPARγ expression was evidenced. It has previously been demonstrated that the PPARγ2 isoform regulates intracellular lipid stores (37), whereas PPARγ1 regulates the expression of inflammatory cytokines through inhibition of nuclear factor (NF)-κB (37). Studies have shown a decreased number of PPARγ-positive hepatocytes (26) and an increased number of hepatocytes expressing NF-κB and tumor necrosis factor (TNF)-α following CCl4 treatment (38). The severity of liver lesions has been found to be correlated with a reduction in the number of hepatocytes expressing PPARγ (38). In addition, results of another study indicated that the PPARγ1 isoform was suppressed while, in contrast, PPARγ2 levels remained unchanged and finally peaked at 8 weeks after CCl4 exposure (37). Considering the results of the present and previous studies, it may be speculated that the decreased expression of PPARγ due to CCl4 toxic activity could be followed by an increased inflammatory response mediated via NF-κB and TNF-α, which leads subsequently to indirect intracellular lipid accumulation and apoptosis. A recovery of PPARγ expression was observed following co-exposure with Bv extract, particularly with nano-encapsulated Bv extract, which may also contribute to the protective effect against CCl4-induced apoptosis, whereas protection of the mitochondrial membrane potential by PPARγ overexpression through the upregulation of anti-apoptotic Bcl-2 family proteins has previously been demonstrated (39).

Formulated and unformulated Bv extract were both efficient against the anti-proliferative and pro-apoptotic actions of CCl4 through suppression of CCl4-induced caspase-3 activation and lipid accumulation. The protective effect was more pronounced following exposure to the formulated β-CD extract compared with the unformulated one, which is probably due to the increased solubility of the complex form, which assured that mitochondrial bioenergetics were balanced.

In conclusion, Bv extract complexed with β-CD protects Huh7 cells against apoptosis and cytotoxicity induced by CCl4 and represents a more potent liver protector against toxic chemical substances and drugs than does the pure extract.

Acknowledgements

The present study was supported by strategic grant POSDRU/159/1.5/S/133391, Project ‘Doctoral and Post-doctoral programs of excellence for highly qualified human resources training for research in the field of Life sciences, Environment and Earth Science’ cofinanced by the European Social Found within the Sectorial Operational Program Human Resources Development 2007–2013.

References

1 

Fallah HH, Zareei MA, Ziai SA, et al: The effects of Taraxacum officinale L. and Berberis vulgaris L. root extracts on carbon tetrachloride induced liver toxicity in rats. J Med Plants. 9:45–52. 2010.

2 

Hermenean A, Popescu C, Ardelean A, Stan M, Hadaruga N, Mihali CV, Costache M and Dinischiotu A: Hepatoprotective effects of Berberis vulgaris L. extract/β cyclodextrin on carbon tetrachloride-induced acute toxicity in mice. Int J Mol Sci. 13:9014–9034. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Weber LW, Boll M and Stampfl A: Hepatotoxicity and mechanism of action of haloalkanes: Carbon tetrachloride as a toxicological model. Crit Rev Toxicol. 33:105–136. 2003. View Article : Google Scholar : PubMed/NCBI

4 

Ruch RJ, Klaunig JE, Schultz NE, Askari AB, Lacher DA, Pereira MA and Goldblatt PJ: Mechanism of chlorophorm and carbon tetrachloride toxicity in primary cultured mouse hepatocytes. Environ Health Perspect. 69:301–305. 1986. View Article : Google Scholar : PubMed/NCBI

5 

Videla LA: Oxidative stress signaling underlying liver disease and hepatoprotective mechanisms. World J Hepatol. 1:72–78. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Balahoroğlu R, Dülger H, Özbek H and Bayram I Şekeroğlu: Protective effects of antioxidants on the experimental liver and kidney toxicity in mice. Eur J Gen Med. 5:157–164. 2008.

7 

Manibusan MK, Odin M and Eastmond DA: Postulated carbon tetrachloride mode of action: A review. J Environ Sci Health C Environ Carcinog Ecotoxicol Rev. 25:185–109. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Arayne MS, Sultana N and Bahadur SS: The Berberis story: Berberis vulgaris in therapeutics. Pak J Pharm Sci. 20:83–92. 2007.PubMed/NCBI

9 

Ivanovska N and Philipov S: Study on the anti-inflamatory action of Berberis vulgaris root extract, alkaloid fractions and pure alkaloids. Int J Immunopharmacol. 18:553–561. 1996. View Article : Google Scholar : PubMed/NCBI

10 

Singh A, Duggal S, Kaur N and Singh J: Berberine. Alkaloid with wide spectrum of pharmacological activities. J Nat Prod (Gorakhpur). 3:64–75. 2010.

11 

Freile ML, Giannini F, Pucci G, Sturniolo A, Rodero L, Pucci O, Balzareti V and Enriz RD: Antimicrobial activity of aqueous extracts and of berberine isolates from Berberis heterophylla. Fitoterapia. 74:702–705. 2003. View Article : Google Scholar : PubMed/NCBI

12 

Iizuka N, Miyamoto K, Okita K, Tangoku A, Hayashi H, Yosino S, Abe T, Morioka T, Hazama S and Oka M: Inhibitory effect of Coptidis rhizoma and berberine on the proliferation of human esophageal cancer cell lines. Cancer Lett. 148:19–25. 2000. View Article : Google Scholar : PubMed/NCBI

13 

Kuo CL, Chi CW and Liu TY: The anti-inflamatory potential of berberine in vitro and in vivo. Cancer Lett. 203:127–137. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Kupeli E, Koşar M, Yeşilada E, Hüsnü K and Başer C: A comparative study on the anti-inflamatory, antinociceptive and antipyretic effects of isoquinoline alkaloids from the root of turkish Berberis species. Life Sci. 72:645–657. 2002. View Article : Google Scholar : PubMed/NCBI

15 

Feng Y, Siu KY, Ye X, Wang N, Yuen MF, Leung CH, Tong Y and Kobayashi S: Hepatoprotective effects of berberine on carbon tetrachloride-induced acute hepatotoxicity in rats. Chin Med. 5:332010. View Article : Google Scholar : PubMed/NCBI

16 

Rasheed A, Kumanr A and Sravahthi V: Cyclodextrins as drug carrier molecule: A review. Sci Pharm. 76:567–598. 2008. View Article : Google Scholar

17 

Guo L, Dial S, Shi L, Branham W, Liu J, Fang JL, Green B, Deng H, Kaput J and Ning B: Similarities and differences in the expression of drug-metabolizing enzymes between human hepatic cell lines and primary human hepatocytes. Drug Metab Dispos. 39:528–538. 2011. View Article : Google Scholar : PubMed/NCBI

18 

Imanshahidi M and Hosseinzadeh H: Pharmacological and therapeutical effects of Berberis vulgaris and its active constituent, berberine. Phytother Res. 22:999–1012. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Zhang BJ, Xu D, Guo Y, Ping J, Chen LB and Wang H: Protection by and anti-oxidant mechanism of berberine against rat liver fibrosis induced by multiple hepatotoxic factors. Clin Exp Pharmacol Physiol. 35:303–309. 2008. View Article : Google Scholar : PubMed/NCBI

20 

Fukuda K, Hibiya Y, Mutoh M, Koshiji M, Akao S and Fujiwara H: Inhibition of berberine of cyclooxygenase-2 transcriptional activity in human colon cancer cells. J Ethnopharmacol. 66:227–233. 1999. View Article : Google Scholar : PubMed/NCBI

21 

Meng Z, Li T, Ma X, Wang X, Van Ness C, Gan Y, Zhou H, Tang J, Lou G, Wang Y, et al: Berbamine inhibits the growth of liver cancer cells and cancer-initiating cells by targeting Ca2+/calmodulin-dependent protein kinase II. Mol Cancer Ther. 12:2067–2077. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Ma S, Wang Y, Shang X, et al: Formulation of berberine hydrochloride and hydroxypropyl-β-cyclodextrin inclusion complex with enhanced dissolution and reduced bitterness. Trop J Pharm Res. 11:871–877. 2012.

23 

Ziolkowski W, Szkatula M, Nurczyk A, Wakabayashi T, Kaczor JJ, Olek RA, Knap N, Antosiewicz J, Wieckowski MR and Wozniak M: Methyl-beta-cyclodextrin induces mitochondrial cholesterol depletion and alters the mitochondrial structure and bioenergetics. FEBS Lett. 584:4606–4610. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Kiss T, Fenyvesi F, Bácskay I, Váradi J, Fenyvesi E, Iványi R, Szente L, Tósaki A and Vecsernyés M: Evaluation of the cytotoxicity of β-cyclodextrin derivatives: Evidences for the role of cholesterol extraction. Eur J Pharm Sci. 40:376–380. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Kamel HH, Azza HA, Walaa MSA and Amira HM: Protective effect of some antioxidants against CCl4-induced toxicity in liver cells from BRL3A cell line. J Am Sci. 6:992–993. 2010.

26 

Guo XL, Liang B, Wang XW, Fan FG, Jin J, Lan R, Yang JH, Wang XC, Jin L and Cao Q: Glycyrrhizic acid attenuates CCl4-induced hepatocyte apoptosis in rats via a p53-mediated pathway. World J Gastroenterol. 19:3781–3791. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Chen J, Tao X, Li L, Sun A, Wang Y and Zhang S: Protective effect of blueberry anthocyanins in a CCl4-induced injury model in human embryonic liver cells. Food Agric Immunol. 25:274–286. 2013. View Article : Google Scholar

28 

Elmore S: Apoptosis: A review of programmed cell death. Toxicol Pathol. 35:495–516. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Saelens X, Festjens N, Van de Walle L, van Gurp M, van Loo G and Vandenabeele P: Toxic proteins released from mitochondria in cell death. Oncogene. 23:2861–2874. 2004. View Article : Google Scholar : PubMed/NCBI

30 

Saraste A and Pulkki K: Morphological and biomedical hallmarks of apoptosis. Cardiovasc Res. 45:528–537. 2000. View Article : Google Scholar : PubMed/NCBI

31 

Aydin HH, Celik A, Deveci R, Karacali S, Saydam G, Omay Bedii S and Batur Y: Induction of apoptosis by fatty acid ethyl esters in HepG2 cells. Food Chem Toxicol. 43:139–145. 2005. View Article : Google Scholar : PubMed/NCBI

32 

Bao W, Pan H, Lu M, Ni Y, Zhang R and Gong X: The apoptotic effect of sarsasapogenin from Anemarrhena asphodeloides on HepG2 human hepatoma cells. Cell Biol Int. 31:887–892. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Fujimoto Y, Itabe H, Kinoshita T, Homma KJ, Onoduka J, Mori M, Yamaguchi S, Makita M, Higashi Y, Yamashita A and Takano T: Involvement of ACSL in local synthesis of neutral lipids in cytoplasmic lipid droplets in human hepatocyte HuH7. J Lipid Res. 48:1280–1292. 2007. View Article : Google Scholar : PubMed/NCBI

34 

Peyrou M, Ramadori P, Bourgoin L and Foti M: PPARs in liver diseases and cancer: Epigenetic regulation by microRNAs. PPAR Res. 2012:7578032012. View Article : Google Scholar : PubMed/NCBI

35 

Rakic B, Sagan SM, Noestheden M, Bélanger S, Nan X, Evans CL, Xie XS and Pezacki JP: Peroxisome proliferator-activated receptor alpha antagonism inhibits hepatitis C virus replication. Chem Biol. 13:23–30. 2006. View Article : Google Scholar : PubMed/NCBI

36 

Toyoda M, Takagi H, Horiguchi N, Kakizaki S, Sato K, Takayama H and Mori M: A ligand for peroxisome proliferator activated receptor gamma inhibits cell growth and induces apoptosis in human liver cancer cells. Gut. 50:563–567. 2002. View Article : Google Scholar : PubMed/NCBI

37 

Morán-Salvador E, Titos E, Rius B, González-Périz A, García-Alonso V, López-Vicario C, Miquel R, Barak Y, Arroyo V and Clària J: Cell-specific PPARγ deficiency establishes anti-inflamatory and anti-fibrogenic properties for this nuclear receptor in non-parenchimal liver cells. J Hepatol. 59:1045–1053. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Orfila C, Lepert JC, Alric L, Carrera G, Béraud M and Pipy B: Immunohistochemical distribution of nuclear factor kappaB and peroxisome proliferated-activated receptors in carbon tetrachloride- induced chronic liver injury in rats. Histochem Cell Biol. 123:585–593. 2005. View Article : Google Scholar : PubMed/NCBI

39 

Wu JS, Lin TN and Wu KK: Rosiglitazone and PPAR-gamma overexpression protect mitochondrial membrane potential and prevent apoptosis by upregulating anti-apoptotic Bcl-2 family proteins. J Cell Physiol. 220:58–71. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2017
Volume 13 Issue 5

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ivan A, Herman H, Balta C, Hadaruga DI, Mihali CV, Ardelean A and Hermenean A: Berberis vulgaris extract/β-cyclodextrin complex increases protection of hepatic cells via suppression of apoptosis and lipogenesis pathways. Exp Ther Med 13: 2143-2150, 2017
APA
Ivan, A., Herman, H., Balta, C., Hadaruga, D.I., Mihali, C., Ardelean, A., & Hermenean, A. (2017). Berberis vulgaris extract/β-cyclodextrin complex increases protection of hepatic cells via suppression of apoptosis and lipogenesis pathways. Experimental and Therapeutic Medicine, 13, 2143-2150. https://doi.org/10.3892/etm.2017.4240
MLA
Ivan, A., Herman, H., Balta, C., Hadaruga, D. I., Mihali, C., Ardelean, A., Hermenean, A."Berberis vulgaris extract/β-cyclodextrin complex increases protection of hepatic cells via suppression of apoptosis and lipogenesis pathways". Experimental and Therapeutic Medicine 13.5 (2017): 2143-2150.
Chicago
Ivan, A., Herman, H., Balta, C., Hadaruga, D. I., Mihali, C., Ardelean, A., Hermenean, A."Berberis vulgaris extract/β-cyclodextrin complex increases protection of hepatic cells via suppression of apoptosis and lipogenesis pathways". Experimental and Therapeutic Medicine 13, no. 5 (2017): 2143-2150. https://doi.org/10.3892/etm.2017.4240