Skip to main content

Proteomic analysis of the inhibitory effect of epigallocatechin gallate on lipid accumulation in human HepG2 cells

Abstract

Background

(−)-Epigallocatechin-3-gallate (EGCG), the most abundant catechin found in green tea, effectively reduces body weight and tissue and blood lipid accumulation. To explore the mechanism by which EGCG inhibits cellular lipid accumulation in free fatty acid (FFA) induced HepG2 cell culture, we investigated the proteome change of FFA-induced HepG2 cells exposed to EGCG using two-dimensional gel electrophoresis and mass spectrometry.

Results

In this study, 36 protein spots showed a significant change in intensity by more than 1.5-fold from the control group to the FFA group and from the FFA group to the FFA + EGCG group. Among them, 24 spots were excised from gels and identified by LC-MS/MS. In total, 18 proteins were successfully identified. All identified proteins were involved in lipid metabolism, glycometabolism, antioxidant defense, respiration, cytoskeleton organization, signal transduction, DNA repair, mRNA processing, iron storage, or were chaperone proteins. This indicated that these physiological processes may play roles in the mechanism of inhibition of lipid accumulation by EGCG in FFA-induced HepG2 cells. Western blotting analysis was used to verify the expression levels of differentially expressed proteins, which agree with the proteomic results.

Conclusions

From the proteomic analysis, we hypothesized that EGCG reduced cellular lipid accumulation in FFA-induced HepG2 cells through the activation of AMP-activated protein kinase (AMPK) resulting from the generation of reactive oxygen species (ROS). The induction of ROS may be a result of EGCG regulation of the antioxidant defense system. Activation of AMPK shifted some FFA toward oxidation, away from lipid and triglyceride storage, and suppressed hepatic gluconeogenesis. The findings of this study improve our understanding of the molecular mechanisms of inhibition of lipid accumulation by EGCG in HepG2 cells.

Background

Nonalcoholic fatty liver disease (NAFLD), defined by excessive liver fat accumulation related to metabolic syndrome, is a leading cause of progressive liver disease. NAFLD is a clinicopathological term that encompasses a disease spectrum ranging from simple lipid accumulation in hepatocytes (hepatic steatosis) to hepatic steatosis with inflammation (steatohepatitis), fibrosis, and cirrhosis [1]. Excess hepatic lipid accumulation is associated with nutritional factors, drugs, and multiple genetic defects in energy metabolism.

Green tea is widely consumed throughout the world, especially in East Asian countries. Research indicates that green tea is beneficial to health and many components of tea have specific health-promoting effects [2, 3]. Studies have suggested that (−)-epigallocatechin-3-gallate (EGCG), the most abundant catechin found in green tea, could effectively reduce body weight and tissue and blood fat accumulation [4, 5]. In high-fat-fed mice, EGCG decreased liver weight, liver triglycerides, plasma alanine aminotransferase concentrations, lipid accumulation in hepatocytes [6], and reduced the development of experimental nonalcoholic steatohepatitis through its effect on lipid metabolism [7]. EGCG treatment could effectively reduce fatty liver incidence, liver damage, and liver triglyceride levels in Male C57BL/6J mice fed with a high-fat, Western-style diet [8]. These beneficial effects of EGCG are associated with decreased lipid absorption and reduced levels of inflammatory cytokines. Previous studies have suggested that EGCG and green tea might modulate expression of lipid metabolism-related genes. EGCG treatment up-regulated several genes related to fat oxidation and thermogenesis, including liver acyl-CoA oxidase (AOX), medium-chain acyl-CoA dehydrogenase (MCAD), muscle uncoupling protein-2 (UCP2) and uncoupling protein-3 (UCP3), and fatty acid translocase [9–11]. Moreover, EGCG treatment down-regulated several genes related to fatty acid synthesis and storage in the liver and white adipose tissue, including acetyl-CoA carboxylase (ACC), fatty acid synthase (FAS), malic enzyme (ME), glucose-6-phosphate dehydrogenase (G6PDH), glycerol-3-phosphate dehydrogenase (G3PDH), and stearoyl-CoA desaturase-1 (SCD1) [12–15].

Despite the amount of work on EGCG, there is little proteomic information available on EGCG inhibiting lipid accumulation in free fatty acid (FFA) induced HepG2 cells. To investigate the inhibitory effect of EGCG on FFA-induced lipid accumulation, HepG2 cells were exposed to FFA co-treated with 50 μM EGCG. The proteome changes of FFA-induced HepG2 cells were investigated by two-dimensional gel electrophoresis (2-DE) combined with matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF/TOF MS). The objective of this study was to obtain an improved understanding of the mechanism by which EGCG inhibits lipid accumulation in FFA-induced HepG2 cells.

Results

Effect of EGCG on FFA-induced intracellular lipid accumulation in HepG2 cells

Fatty liver results from an imbalance between lipid availability and lipid metabolism. Palmitic and oleic acids are the most abundant FFA in liver triglycerides in both normal subjects and patients with NAFLD [16]. Exposure of HepG2 cells to exogenous FFA leads to significant intracellular lipid accumulation [17]. HepG2 cells loaded with 1 mM FFA mixture (oleic acid [OA] and palmitic acid [PA], 2:1) mimics benign chronic steatosis in humans [18]. As shown in Figure 1A, the intracellular lipid content could be significantly lowered by treatment with 50 μM EGCG. This suggested that EGCG could significantly inhibit FFA-induced intracellular lipid accumulation in HepG2 cells. This result was also confirmed by the quantification of intracellular triglycerides and cholesterol contents. In Figure 1B, EGCG treatment showed significantly lower triglyceride levels (p ≤ 0.05). The content of cholesterol was also lowered but not significantly. The effects of EGCG on FFA-stimulated HepG2 cell viability were examined by MTT assay and annexin V staining. The results showed that cell viability was not compromised by 50 μM EGCG treatment after 24 h of exposure (Figure 1C and D).

Figure 1
figure 1

EGCG reduced FFA-induced intracellular lipid accumulation in HepG2 cells. Lipid content detected by a quantitative Oil Red O dye method (A), Triglyceride and cholesterol levels in FFA-overloaded HepG2 cells in different treatment groups (B), The effect of EGCG on FFA-overloaded HepG2 cells. Proliferation and apoptosis were assessed by MTT assay (C) and PI/annexin V staining (D).

Change in protein abundance in the three groups

To explore the inhibitory effect of EGCG on FFA-induced lipid accumulation in HepG2 cells, the proteomes of the control, FFA-induced (FFA) and FFA-induced co-treated with 50 μM EGCG groups (FFA + EGCG) were analyzed by 2-DE (Figure 2). Among the tested samples, more than 800 protein spots were reproducibly detected with PDQuest 8.0.1 software on Coomassie Brilliant Blue (CBB) G-250-stained gels. The control and FFA + EGCG groups had 142 and 151 protein spots that showed a significant change in expression level in when compared with the FFA group, respectively. Moreover, 36 protein spots showed a significant change in intensity by more than 1.5-fold from the control group to the FFA group and from the FFA group to the FFA + EGCG group. Among them, 24 spots that changed at least 2.0-fold were excised from gels and identified by MALDI-TOF/TOF MS. In total, 18 protein spots were successfully identified and the results are summarized in Table 1. The functions of the differentially expressed proteins were obtained using their protein accession numbers from the SwissProt/NCBI protein function summary. All identified proteins in this study were involved in multiple functional groups (Table 1), including lipid metabolism, glycometabolism, antioxidant defense, respiration, cytoskeleton organization, signal transduction, DNA repair, mRNA processing, and iron storage, or were chaperone proteins.

Figure 2
figure 2

Comparison of 2-D electrophoresis analysis of the proteins expressed in FFA-stimulated HepG2 cells derived from different treatment groups. Control (A), FFA (B), FFA + EGCG (C). Proteins that exhibited a significant expression change (≥ 1.5-fold, p ≤ 0.05) from the control group to FFA group while from the FFA group to FFA + EGCG group are labeled in the figures.

Table 1 Differentially expressed proteins identified by MS or MS/MS

Validation of differentially expressed proteins by western blotting

Western blotting analysis was performed in triplicate to confirm the differentially expression proteins found in the proteomic analysis. FFA-induced HepG2 cells were treated with 50 μM EGCG for 24 h. Equal amounts of total proteins from different treated cells were used for western blotting analysis. The results (Figure 3) suggested that the expressed levels of peroxiredoxin-6 (Prdx6) and galactokinase (GALK) were significantly lower (p ≤ 0.05) and succinate dehydrogenase flavoprotein subunit (SDHA) was significantly higher (p ≤ 0.05) in the FFA + EGCG group compared with the FFA group. Therefore, the western blotting results agree with the proteomic results.

Figure 3
figure 3

Validation of Prdx6, GALK, and SDHA expression levels in HepG2 cells by Western blotting. The expression levels of Prdx6, GALK and SDHA in the control, FFA and FFA + EGCG groups were detected by Western blotting. β-actin was used as a loading control. Similar results were found in the three independent experiments and the representative results are shown.

Discussion

Lipid accumulation is the main cause of NAFLD, which results in fatty tissue degeneration [1, 19]. This lipid accumulation in hepatocytes results from an imbalance between lipogenesis and lipolysis metabolism causing eventual lipoperoxidative stress and hepatic injury [20]. Previous studies suggest that the inhibitory action of EGCG on lipid accumulation is mediated via the AMP-activated protein kinase (AMPK) pathway [21–23]. Activation of AMPK can induce ATP generation through glycolysis and β-oxidation, and suppress fatty acid and cholesterol syntheses, and gluconeogenesis [24]. However, the inhibitory effect of EGCG on FFA-induced intracellular lipid accumulation at the proteomic level has not been investigated. In this study, we reported a comprehensive proteome, which deduced the target genes of EGCG as well as its inhibitory effect on lipid accumulation in FFA-induced HepG2 cells. Eighteen differentially expressed proteins were successfully identified. These proteins are involved in lipid metabolism, glycometabolism, antioxidant defense, respiration, cytoskeleton organization, signal transduction, DNA repair, mRNA processing, and iron storage, or are chaperone proteins. This indicates that these physiological processes may play a role in the mechanism by which EGCG inhibits cellular lipid accumulation in FFA-induced HepG2 cells.

Proteins involved in redox regulation and energy metabolism

After treatment with EGCG, two differentially expressed proteins related to redox and respiratory regulation were identified in HepG2 cells. Prdx6 is the sixth mammalian member of the peroxiredoxin family, which has an important role in antioxidant defense [25, 26]. Several studies reported that an increase in reactive oxygen species (ROS) could lead to AMPK activation [27, 28]. EGCG significantly induced generation of ROS and reduced glutathione (GSH) in cells [21–23]. AMPK is thought to be a novel target for the treatment of obesity and type II diabetes [29]. Activation of AMPK in liver and skeletal muscle leads to the stimulation of fatty acid oxidation and inhibition of lipogenesis, glucose production and protein synthesis [30, 31]. However, these effects could be eliminated by ROS scavenger, N-acetylcysteine, treatment [22, 23, 28]. In our proteomic experiment, Prdx6 was down-regulated by EGCG treatment. This may be a reason for the increasing ROS levels that lead to AMPK activation, preventing lipid accumulation in cells. Succinate dehydrogenase (SDH or ubiquinone) is an enzyme that controls the transcription of metabolism-related genes in mitochondria and promotes metabolism of glucose and lipids [32, 33]. SDH is related to oxidative metabolism and is an indicator of oxidative capacity in cells [34]. Prior studies have found that a reduction in oxidative enzymes is correlated with a reduced capacity for lipid oxidation and increased risk for obesity [35, 36]. The mRNA expression levels of SDH are reduced in mice with type II diabetes mellitus and obesity [37]. Several studies have also shown that weight loss and exercise could result in a significant increase in SDH activity [38, 39]. Our proteomic analysis showed that SDHA was up-regulated by EGCG treatment in FFA-induced HepG2 cells, indicating that EGCG increased lipid oxidation by enhancing energy expenditure and fat oxidation in the mitochondria of HepG2 cells.

Proteins involved in lipid metabolism

Proteins involved in lipid metabolism were changed significantly by EGCG treatment in HepG2 cells. Platelet-activating factor acetylhydrolase (PAFAH), a unique member of the phospholipase A2 superfamily, is characterized by its ability to specifically hydrolyze platelet-activating factor (PAF) and glycerophospholipids [40]. PAF is a potent pro-inflammatory phospholipid that activates cells involved in inflammation and stimulates ROS generation [41–43]. Down-regulation of platelet-activating factor acetylhydrolase IB subunit gamma (PAFAH1B3) by EGCG treatment might lead to an increase of PAF, which could stimulate the generation of ROS and activating AMPK in HepG2 cells. Short/branched-chain acyl-CoA dehydrogenase (SBCAD) catalyzes the first step in the mitochondrial β-oxidation of L-2-methylated short acyl-CoA compounds [44]. SBCAD deficiency is characterized by accumulation of 2-methylbutyrylglycine (2MBG), which induced an increase of lipid oxidation and a decrease of antioxidant defenses (decreased GSH) in rat [45]. In our proteomic experiment, SBCAD was down-regulated in the EGCG-treated group. This might result in a decrease of antioxidant defense and a high level of cellular ROS, which could activate AMPK to prevent cellular lipid accumulation.

Proteins involved in glycometabolism

Two proteins involved in glycometabolism were down-regulated by EGCG treatment. GALK catalyzes the phosphorylation of α-galactose to galactose-1-phosphate in the second step of the Leloir pathway, a metabolic pathway found in most organisms for the catabolism of β-galactose to glucose-1-phosphate [46]. Glucose-1-phosphate could be metabolized to glucose-6-phosphate providing substrate for the pentose phosphate pathway, which generates ribose-5-phosphate and NADPH for the biosynthesis of fatty acids and sterols [47]. A study reported that unsaturated fatty acids could increase the activation of galactokinase and galactose-1-phosphate uridyltransferase, and stimulate sterol biosynthesis from galactose [48]. Down-regulation of galactokinase reduces ribose-5-phosphate and NADPH from the pentose phosphate pathway, resulting in a reduction of the biosynthesis of fatty acids and sterols in FFA-induced HepG2 cells. Lysosomal protective protein (PPCA) appears to be essential for the activity of β-galactosidase, which catalyzes the hydrolysis of β-galactosides into β-galactose and glucoside. PPCA associates with β-galactosidase and exerts a protective function necessary for its stability and activity [49]. Down-regulation of PPCA could reduce the activity of β-galactosides and decrease β-galactose for the Leloir pathway, inhibiting the biosynthesis of fatty acids and sterols from galactose [47, 48].

Proteins involved in biological regulation and signal transduction

Some proteins involved in biological regulation and signal transduction were also identified in HepG2 cells, including serine/threonine protein phosphatase-alpha catalytic subunit (PP-1A), phosphoserine phosphatase (PSP), 14-3-3 protein gamma and prohibitin (PHB). PP-1A and PSP belong to the phosphoprotein phosphatase family, which remove the phosphate from the serine or threonine residues of phosphoproteins [50]. Previous study reported that a glucose-induced activation of the transcription of the FAS gene was markedly reduced by okadaic acid, an inhibitor of protein serine/threonine phosphatases 1 (PP1) and PP2A, and by AICAR, a cell-permeable activator of the AMPK [51]. These results indicated that the reduction of the FAS gene involves a phosphorylation/dephosphorylation mechanism and AMPK activation. Therefore, down-regulation of PP-1A and PSP might indicate an inhibition of lipid biosynthesis and an activation of AMPK in HepG2 cells by EGCG treatment. 14-3-3 protein gamma belongs to the 14-3-3 protein family, which has the ability to bind many functionally diverse signaling proteins, including kinases, phosphatases, and transmembrane receptors [52, 53]. The transcriptional co-activator transducer of regulated CREB activity 2 (TORC2) is a pivotal component of gluconeogenesis [54]. Phosphorylated TORC2 is sequestered in the cytoplasm via a phosphorylation-dependent interaction with 14-3-3 proteins and degraded by 26S proteasome to inhibit the gluconeogenic program [55]. A report suggested that inhibition of enhanced gluconeogenesis induced by high-fat and high-fructose diet could improve lipid metabolism and hepatic steatosis in mice [56]. Thus, up-regulation of 14-3-3 protein by EGCG treatment in FFA-induced HepG2 cells might indicate that the hepatic steatosis induced by overloaded FFA was improved through inhibition of gluconeogenesis. PHB comprise two evolutionarily conserved proteins, prohibitin-1 (PHB1) and prohibitin-2 (PHB2), and are present in a high molecular-weight complex in the inner membrane of mitochondria [57, 58]. PHB1 decreased insulin-stimulated oxidation of glucose and fatty acids, implying that PHB1 may play a role in promoting fat accumulation [59]. A microarray analysis showed that the expression levels of PHB were increased during 3T3-L1 cell adipogenesis [60]. Several studies showed that depletion of PHB1 or PHB2 in C. elegans or 3T3-L1 cells could significantly decrease adipose accumulation and adipogenesis [61]. In the current study, EGCG decreased the expression of PHB, which might be one of the reasons that EGCG could inhibit lipid accumulation in FFA-induced HepG2 cells.

Conclusions

In summary, this study demonstrates that EGCG can significantly suppress the lipid accumulation in FFA-induced HepG2 cells. Using a proteomic approach, we identified 18 differentially expressed proteins responsive to EGCG treatment involving multiple cellular processes. From the proteomic analysis, we supposed that EGCG reduced cellular lipid accumulation in FFA-induced HepG2 cells through the activation of AMPK resulting from the generation of ROS. The induction of ROS may be because EGCG regulated the antioxidant defense system in HepG2 cells (Figure 4). Activation of AMPK shifted some FFA toward oxidation, away from lipid and triglyceride storage, and suppressed hepatic gluconeogenesis in HepG2 cells. Findings of this study provide information to improve our understanding of the molecular mechanisms of the inhibition of lipid accumulation by EGCG in HepG2 cells.

Figure 4
figure 4

A simple model of the proposed mechanism by which EGCG inhibits lipid accumulation in FFA-induced HepG2 cells. Arrowheads indicate the direct or indirect interactions. The up-regulated proteins are marked with red and the down-regulated proteins are marked with blue.

Methods

Materials

All chemicals used were of analytic grade. EGCG was obtained from Sigma (St. Louis, MO, USA), the purity of EGCG was ≥95%. Oleic acid and palmitic acid were also obtained from Sigma. Antibodies to β-actin and Prdx6 were obtained from GeneTex (Irvine, CA, USA). Antibodies to SDHA and GALK were obtained from Abcam (Cambridge, UK).

Cell culture and EGCG stimulation

Experiments were approved by the ethics committee of Tsinghua University. HepG2 cells (human liver hepatocellular carcinoma cell line) were obtained from the cell bank of the Type Culture Collection of Chinese Academy of Sciences, and were cultured in DMEM medium (Thermo, South Logan, UT, USA) with 10% fetal calf serum, 100 U/mL penicillin, 100 μg/mL streptomycin, and 2 mM L-glutamine and kept at 37°C in a humidified atmosphere (Nuaire, Plymouth, MN, USA) with 5% CO2. To induce fat-overloading, HepG2 cells were exposed to 1 mM of FFA mixture (OA / PA, 2:1) mixed with 1% FFA-free bovine serum albumin [17, 28, 62]. Stock solutions of 50 mM FFA were prepared as reported previously [63]. When the cells reached 70% confluence, they were incubated in serum-free medium for 24 h before treatments. Then, the cells were stimulated with 1 mM FFA and co-treated with EGCG for 24 h.

Cell proliferation and apoptosis assay

The effect of EGCG on cell proliferation was assessed using MTT assay as previously reported [64]. Briefly, HepG2 cells were plated in 96-well plates (5 × 103 cells/well) and treated with 1 mM FFA with various concentrations of EGCG for 24 h. Then cells were incubated with 100 μL MTT solution (0.5 mg/mL MTT in PBS buffer) for 4 h. The absorbance was measured with a Multiskan MK3 (Thermo) at 490 nm. Apoptotic cells were detected using flow cytometry with PI/annexin V staining [64]. Cells were seeded in six-well plates (4 × 106 cells/well) and co-incubated with 0, 50, and 100 μM of EGCG and 1 mM FFA for 24 h. After harvesting with 0.25% trypsin, cells were resuspended with 200 μL binding buffer and incubated with 10 μL Annexin V-FITC and 5 μL PI for 15 min at room temperature. Sample solutions were adjusted to 500 μL using binding buffer and analyzed with a FACScan flow cytometer (Becton Dickinson Biosciences, San Jose, CA, USA) within an hour of sample preparation. The data were analyzed by Modfit3.0 (Verity Software House, Topsham, ME, USA). All treatments were performed in triplicate.

Oil Red O staining

The Oil Red O (ORO) staining was according to Amacher et al. [65, 66]. Briefly, the cells were washed twice with ice-cold PBS and fixed with 10% formalin for 1 h, and stained with Oil Red O solution for 2 h at room temperature. After staining, cells were washed twice with distilled water to remove unbound dye. To quantitate the intracellular lipid content levels, isopropanol was added to each sample shaken at room temperature for 5 min, and samples were read spectrophotometrically at 520 nm.

Triglycerides and cholesterol assay

For lipidic determinations, homogenates from cells were extracted according to a Heider et al. [67]. Briefly, each sample was homogenized with isopropyl alcohol. The resulting mixture was shaken at room temperature for 1 h and then centrifuged at 1200 × g for 10 min. The supernatant was collected for analysis of hepatic TG and TC content. The residue was dissolved in 0.1 M sodium hydroxide and an aliquot was taken for protein determination with an RC DC Protein Assay Kit (Bio-Rad, Hercules, CA, USA). Triacylglycerol and cholesterol were measured using enzymatic method kits (Cell Biolabs, Inc. San Diego, CA, USA) according to the manufacturer’s instructions.

Protein extract and sample preparation for 2-DE

The HepG2 cells were harvested by trypsinization and washed twice with ice-cold PBS buffer. The cells were lysed in lysis buffer (7 M urea, 2 M thiourea, 4% [w/v] CHAPS, 65 mM DTT, 2% [v/v] Bio-Lyte pH [3–10], 2% protease inhibitor cocktail), mixed by vortexing, kept in an ice bath for 2 h, then sonicated in an ice bath. The sample was clarified by centrifugation at 15000 × g for 1 h at 4°C, and the supernatants stored at −80°C until use for 2-DE. Protein content was quantified using the RC DC Protein Assay Kit (Bio-Rad).

2-DE and image analysis

About 1.3 mg protein dissolved in 350 μL rehydration buffer was applied to IPG strips (17 cm, pH 3–10, Bio-Rad), which was allowed to rehydrate for 13 h at 50 V (20°C). Subsequently, isoelectric focusing (IEF) was performed at using a Protean IEF Cell (Bio-Rad) under the following conditions: 250 V for 1 h with a slow increase in voltage, 500 V for 1 h with a slow increase in voltage, 1000 V for 1 h with a slow increase in voltage, 10000 V for 5 h with a linear increase in voltage, and maintained at 10000 V until 60000 Volt-hours (Vh) was reached. After IEF, the strips were equilibrated for 15 min in equilibration buffer I (0.375 M Tris–HCl pH 8.8, 6 M urea, 2% SDS, 20% glycerol, 1% DTT), then re-equilibrated in buffer II containing 2.5% iodoacetamide instead of DTT for 15 min. The strips were transferred onto 12% polyacrylamide gels for SDS-PAGE. Electrophoresis was performed using the PROTEAN II xi Cell system (Bio-Rad) at 10 mA per gel for 30 min, followed by 30 mA until the bromophenol blue marker reached the end of the gel. Gels were run in triplicate for each sample. The gels were stained with modified colloidal CBB G-250 [68] and were scanned using Quantity One 4.6.9 (Bio-Rad) with a Bio-Rad GS800 scanner. Image and statistical analysis was performed with PDQuest 8.0.1 (Bio-Rad) as previously reported [69]. In the quantitative analysis, 1.5 and 0.5 were chosen as the upper and lower limits, respectively. Student’s t-test and a significance level of 95% were used for the statistical analysis of the gels. Each sample was performed in triplicate gels.

Protein in-gel digestion and identification

Protein spots were manually excised from the gel, and digested as previously reported [69]. Mass spectrometric analysis was performed with an Ultraflex MALDI TOF/TOF mass spectrometer (Bruker Daltonik, Bremen, Germany), under the control of FlexControl™ 2.2 software (Bruker Daltonik GmbH). The TOF spectra were recorded in the positive ion reflector mode in a mass range from 800–4000 Da. Ten subspectra with 30 shots per subspectrum were accumulated to generate one main TOF spectrum. After automated assessment of the search results with MASCOT software (Matrix Science, London, UK), the samples not unambiguously identified by PMF were automatically submitted to MS/MS analysis using the LIFT technology in TOF/TOF. A maximum of three strongest peaks of the TOF MS spectrum per sample were chosen for MS/MS analysis. Three subspectra with 50 shots per subspectrum for precursor ions and 15 subspectra with 50 shots per subspectrum for fragment ions were accumulated to produce one main MS/MS spectrum. After the automated analysis, remaining unidentified samples were manually analyzed.

Database searching

Protein identification from MS/MS sequencing spectra was accomplished using the MASCOT database search engine (Matrix Science, London, UK). The searching parameters were set as follows: taxonomy, Homo sapiens; database, NCBInr/Swiss-Prot; enzyme, trypsin; fixed modifications, carbamidomethyl (C); variable modifications, oxidation (M); no restrictions on protein mass; allow up to one missed cleavages. Mass values, Monoisotopic; Peptide Mass Tolerance was set as ±50 ppm; Fragment Mass Tolerance was set as ±0.5 Da. Positive protein identification was based on standard MASCOT criteria for statistical analysis of the LC-MS/MS data. The peptide assignments in the database search results were manually inspected for validation.

Western blotting analysis

The HepG2 cells were harvested by trypsinization and washed twice with ice-cold PBS buffer. The cells were lysed in RIPA buffer (25 mM Tris–HCl pH 7.6, 150 mM NaCl, 1% NP-40, 1% sodium deoxycholate, 0.1% SDS, 2% protease inhibitor cocktail). Equal amounts of protein were separated on a 12% SDS-polyacrylamide gel, blotted onto a PVDF membrane, which was blocked for 1 h at 25°C with 5% wt/vol BSA/TBST (10 mM Tris HCl, pH 7.4, 140 mM NaCl, 0.1% Tween-20) and then incubated with the primary antibody for Prdx6 (1:3000), SDHA (1:1000) and GALK (1:1000) at 4°C overnight. After washing with TBST, the membranes were incubated with the appropriate secondary antibodies for 1 h at 37°C and detected by immuno-staining. After the membranes were scanned, the signal intensity of each band was determined using FluorChem FC2 (Alpha Innotech Co., Ltd, San Leandro, CA, USA).

Statistical analysis

All results were expressed as mean ± SD and analyzed by student’s t-test. A p ≤ 0.05 was considered to be statistically significant.

Abbreviations

EGCG:

(−)-epigallocatechin-3-gallate

2-DE:

Two-dimensional gel electrophoresis

MALDI-TOF/TOF MS:

Matrix-assisted laser desorption ionization time-of-flight mass spectrometry

FFA:

Free fatty acid

OA:

Oleic acid

PA:

Palmitic acid

TG:

Triglycerides

TC:

Cholesterol

AMPK:

AMP-activated protein kinase

Prdx6:

Peroxiredoxin-6

ROS:

Reactive oxygen species

SDH:

Succinate dehydrogenase

SDHA:

Succinate dehydrogenase flavoprotein subunit

PAFAH:

Platelet-activating factor acetylhydrolase

PAF:

Platelet-activating factor

PAFAH1B3:

Platelet-activating factor acetylhydrolase IB subunit gamma

SBCAD:

Short/branched-chain acyl-CoA dehydrogenases

2MBG:

2-methylbutyrylglycine

GSH:

Glutathione

GALK:

Galactokinase

PPCA:

Lysosomal protective protein

PP-1A:

Serine/threonine protein phosphatase-alpha catalytic subunit

PSP:

Phosphoserine phosphatase

FAS:

Fatty acid synthase

TORC2:

Transducer of regulated CREB activity 2

PHB:

Prohibitin.

References

  1. Angulo P, Lindor KD: Non-alcoholic fatty liver disease. J Gastroenterol Hepatol 2002, 17: S186-S190.

    Article  PubMed  Google Scholar 

  2. Suzuki Y, Miyoshi N, Isemura M: Health-promoting effects of green tea. Proc Jpn Acad Ser B Phys Biol Sci 2012, 88: 88–101. 10.2183/pjab.88.88

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  3. Yang CS, Landau JM: Effects of tea consumption on nutrition and health. J Nutr 2000, 130: 2409–2412.

    CAS  PubMed  Google Scholar 

  4. Kao YH, Chang HH, Lee MJ, Chen CL: Tea, obesity, and diabetes. Mol Nutr Food Res 2006, 50: 188–210. 10.1002/mnfr.200500109

    Article  CAS  PubMed  Google Scholar 

  5. Wolfram S, Wang Y, Thielecke F: Anti-obesity effects of green tea: from bedside to bench. Mol Nutr Food Res 2006, 50: 176–187. 10.1002/mnfr.200500102

    Article  CAS  PubMed  Google Scholar 

  6. Bose M, Lambert JD, Ju J, Reuhl KR, Shapses SA, Yang CS: The major green Tea polyphenol, (−)-epigallocatechin-3-gallate, inhibits obesity, metabolic syndrome, and fatty liver disease in high-Fat–Fed mice. J Nutr 2008, 138: 1677–1683.

    CAS  PubMed Central  PubMed  Google Scholar 

  7. Kuzu N, Bahcecioglu IH, Dagli AF, Ozercan IH, Ustündag B, Sahin K: Epigallocatechin gallate attenuates experimental non-alcoholic steatohepatitis induced by high fat diet. J Gastroenterol Hepatol 2008, 23: e465-e470. 10.1111/j.1440-1746.2007.05052.x

    Article  CAS  PubMed  Google Scholar 

  8. Chen YK, Cheung C, Reuhl KR, Liu AB, Lee MJ, Lu YP, Yang CS: Effects of green tea polyphenol (−)-epigallocatechin-3-gallate on newly developed high-fat/Western-style diet-induced obesity and metabolic syndrome in mice. J Agric Food Chem 2011, 59: 11862–11871. 10.1021/jf2029016

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  9. Klaus S, Pultz S, Thone-Reineke C, Wolfram S: Epigallocatechin gallate attenuates diet-induced obesity in mice by decreasing energy absorption and increasing fat oxidation. Int J Obes (Lond) 2005, 29: 615–623. 10.1038/sj.ijo.0802926

    Article  CAS  Google Scholar 

  10. Tokimitsu I: Effects of tea catechins on lipid metabolism and body fat accumulation. Biofactors 2004, 22: 141–143. 10.1002/biof.5520220127

    Article  CAS  PubMed  Google Scholar 

  11. Murase T, Haramizu S, Shimotoyodome A, Nagasawa A, Tokimitsu I: Green tea extract improves endurance capacity and increases muscle lipid oxidation in mice. Am J Physiol Regul Integr Comp Physiol 2005, 288: R708-R715.

    Article  CAS  PubMed  Google Scholar 

  12. Wolfram S, Raederstorff D, Wang Y, Teixeira SR, Elste V, Weber P: TEAVIGO (epigallocatechin gallate) supplementation prevents obesity in rodents by reducing adipose tissue mass. Ann Nutr Metab 2005, 49: 54–63. 10.1159/000084178

    Article  CAS  PubMed  Google Scholar 

  13. Brusselmans K, De Schrijver E, Heyns W, Verhoeven G, Swinnen JV: Epigallocatechin-3-gallate is a potent natural inhibitor of fatty acid synthase in intact cells and selectively induces apoptosis in prostate cancer cells. Int J Cancer 2003, 106: 856–862. 10.1002/ijc.11317

    Article  CAS  PubMed  Google Scholar 

  14. Yeh CW, Chen WJ, Chiang CT, Lin-Shiau SY, Lin JK: Suppression of fatty acid synthase in MCF-7 breast cancer cells by tea and tea polyphenols: a possible mechanism for their hypolipidemic effects. Pharmacogenomics J 2003, 3: 267–276.

    CAS  PubMed  Google Scholar 

  15. Mochizuki M, Hasegawa N: Stereospecific effects of catechin isomers on insulin induced lipogenesis in 3T3-L1 cells. Phytother Res 2004, 18: 449–450. 10.1002/ptr.1437

    Article  CAS  PubMed  Google Scholar 

  16. Araya J, Rodrigo R, Videla LA, Thielemann L, Orellana M, Pettinelli P, Poniachik J: Increase in long-chain polyunsaturated fatty acid n - 6/n - 3 ratio in relation to hepatic steatosis in patients with non-alcoholic fatty liver disease. Clin Sci (Lond) 2004, 106: 635–643. 10.1042/CS20030326

    Article  CAS  Google Scholar 

  17. Gómez-Lechón MJ, Donato MT, Martínez-Romero A, Jiménez N, Castell JV, O’Connor J-E: A human hepatocellular in vitro model to investigate steatosis. Chemico-Biol Interact 2007, 165: 106–116. 10.1016/j.cbi.2006.11.004

    Article  Google Scholar 

  18. Gomez-Lechon MJ, Donato MT, Martinez-Romero A, Jimenez N, Castell JV, O’Connor JE: A human hepatocellular in vitro model to investigate steatosis. Chem Biol Interact 2007, 165: 106–116. 10.1016/j.cbi.2006.11.004

    Article  CAS  PubMed  Google Scholar 

  19. Marchesini G, Brizi M, Bianchi G, Tomassetti S, Bugianesi E, Lenzi M, McCullough AJ, Natale S, Forlani G, Melchionda N: Nonalcoholic fatty liver disease: a feature of the metabolic syndrome. Diabetes 2001, 50: 1844–1850. 10.2337/diabetes.50.8.1844

    Article  CAS  PubMed  Google Scholar 

  20. Musso G, Gambino R, Cassader M: Recent insights into hepatic lipid metabolism in non-alcoholic fatty liver disease (NAFLD). Prog Lipid Res 2009, 48: 1–26. 10.1016/j.plipres.2008.08.001

    Article  CAS  PubMed  Google Scholar 

  21. Hwang JT, Park IJ, Shin JI, Lee YK, Lee SK, Baik HW, Ha J, Park OJ: Genistein, EGCG, and capsaicin inhibit adipocyte differentiation process via activating AMP-activated protein kinase. Biochem Biophys Res Commun 2005, 338: 694–699. 10.1016/j.bbrc.2005.09.195

    Article  CAS  PubMed  Google Scholar 

  22. Moon HS, Chung CS, Lee HG, Kim TG, Choi YJ, Cho CS: Inhibitory effect of (−)-epigallocatechin-3-gallate on lipid accumulation of 3T3-L1 cells. Obesity (Silver Spring) 2007, 15: 2571–2582. 10.1038/oby.2007.309

    Article  CAS  Google Scholar 

  23. Wang CT, Chang HH, Hsiao CH, Lee MJ, Ku HC, Hu YJ, Kao YH: The effects of green tea (−)-epigallocatechin-3-gallate on reactive oxygen species in 3T3-L1 preadipocytes and adipocytes depend on the glutathione and 67 kDa laminin receptor pathways. Mol Nutr Food Res 2009, 53: 349–360. 10.1002/mnfr.200800013

    Article  CAS  PubMed  Google Scholar 

  24. Hwang J-T, Kwon DY, Yoon SH: AMP-activated protein kinase: a potential target for the diseases prevention by natural occurring polyphenols. New Biotechnol 2009, 26: 17–22. 10.1016/j.nbt.2009.03.005

    Article  CAS  Google Scholar 

  25. Pak JH, Manevich Y, Kim HS, Feinstein SI, Fisher AB: An antisense oligonucleotide to 1-cys peroxiredoxin causes lipid peroxidation and apoptosis in lung epithelial cells. J Biol Chem 2002, 277: 49927–49934. 10.1074/jbc.M204222200

    Article  CAS  PubMed  Google Scholar 

  26. Wang Y, Feinstein SI, Fisher AB: Peroxiredoxin 6 as an antioxidant enzyme: protection of lung alveolar epithelial type II cells from H2O2-induced oxidative stress. J Cell Biochem 2008, 104: 1274–1285. 10.1002/jcb.21703

    Article  CAS  PubMed  Google Scholar 

  27. Emerling BM, Weinberg F, Snyder C, Burgess Z, Mutlu GM, Viollet B, Budinger GRS, Chandel NS: Hypoxic activation of AMPK is dependent on mitochondrial ROS but independent of an increase in AMP/ATP ratio. Free Radic Biol Med 2009, 46: 1386–1391. 10.1016/j.freeradbiomed.2009.02.019

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  28. Lin CL, Huang HC, Lin JK: Theaflavins attenuate hepatic lipid accumulation through activating AMPK in human HepG2 cells. J Lipid Res 2007, 48: 2334–2343. 10.1194/jlr.M700128-JLR200

    Article  CAS  PubMed  Google Scholar 

  29. Carling D, Thornton C, Woods A, Sanders MJ: AMP-activated protein kinase: new regulation, new roles? Biochem J 2012, 445: 11–27. 10.1042/BJ20120546

    Article  CAS  PubMed  Google Scholar 

  30. Fediuc S, Gaidhu MP, Ceddia RB: Regulation of AMP-activated protein kinase and acetyl-CoA carboxylase phosphorylation by palmitate in skeletal muscle cells. J Lipid Res 2006, 47: 412–420.

    Article  CAS  PubMed  Google Scholar 

  31. Viollet B, Foretz M, Guigas B, Horman S, Dentin R, Bertrand L, Hue L, Andreelli F: Activation of AMP-activated protein kinase in the liver: a new strategy for the management of metabolic hepatic disorders. J Physiol 2006, 574: 41–53. 10.1113/jphysiol.2006.108506

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  32. Oyedotun KS, Lemire BD: The quaternary structure of the Saccharomyces cerevisiae succinate dehydrogenase. Homology modeling, cofactor docking, and molecular dynamics simulation studies. J Biol Chem 2004, 279: 9424–9431. 10.1074/jbc.M311876200

    Article  CAS  PubMed  Google Scholar 

  33. Rutter J, Winge DR, Schiffman JD: Succinate dehydrogenase - assembly, regulation and role in human disease. Mitochondrion 2010, 10: 393–401. 10.1016/j.mito.2010.03.001

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  34. Goodpaster BH, He J, Watkins S, Kelley DE: Skeletal muscle lipid content and insulin resistance: evidence for a paradox in endurance-trained athletes. J Clin Endocrinol Metab 2001, 86: 5755–5761. 10.1210/jc.86.12.5755

    Article  CAS  PubMed  Google Scholar 

  35. Simoneau JA, Veerkamp JH, Turcotte LP, Kelley DE: Markers of capacity to utilize fatty acids in human skeletal muscle: relation to insulin resistance and obesity and effects of weight loss. FASEB J 1999, 13: 2051–2060.

    CAS  PubMed  Google Scholar 

  36. Sun G, Ukkola O, Rankinen T, Joanisse DR, Bouchard C: Skeletal muscle characteristics predict body fat gain in response to overfeeding in never-obese young men. Metab Clin Exp 2002, 51: 451–456. 10.1053/meta.2002.31324

    Article  CAS  PubMed  Google Scholar 

  37. Yechoor VK, Patti ME, Saccone R, Kahn CR: Coordinated patterns of gene expression for substrate and energy metabolism in skeletal muscle of diabetic mice. Proc Natl Acad Sci U S A 2002, 99: 10587–10592. 10.1073/pnas.142301999

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  38. Kern PA, Simsolo RB, Fournier M: Effect of weight loss on muscle fiber type, fiber size, capillarity, and succinate dehydrogenase activity in humans. J Clin Endocrinol Metab 1999, 84: 4185–4190. 10.1210/jc.84.11.4185

    CAS  PubMed  Google Scholar 

  39. Menshikova EV, Ritov VB, Ferrell RE, Azuma K, Goodpaster BH, Kelley DE: Characteristics of skeletal muscle mitochondrial biogenesis induced by moderate-intensity exercise and weight loss in obesity. J Appl Physiol 2007, 103: 21–27. 10.1152/japplphysiol.01228.2006

    Article  CAS  PubMed  Google Scholar 

  40. Stafforini DM, McIntyre TM, Zimmerman GA, Prescott SM: Platelet-activating factor acetylhydrolases. J Biol Chem 1997, 272: 17895–17898. 10.1074/jbc.272.29.17895

    Article  CAS  PubMed  Google Scholar 

  41. Tjoelker LW, Wilder C, Eberhardt C, Stafforini DM, Dietsch G, Schimpf B, Hooper S, Le Trong H, Cousens LS, Zimmerman GA, et al.: Anti-inflammatory properties of a platelet-activating factor acetylhydrolase. Nature 1995, 374: 549–553. 10.1038/374549a0

    Article  CAS  PubMed  Google Scholar 

  42. Vittos O, Toana B, Vittos A, Moldoveanu E: Lipoprotein-associated phospholipase A2 (Lp-PLA2): a review of its role and significance as a cardiovascular biomarker. Biomarkers 2012, 17: 289–302. 10.3109/1354750X.2012.664170

    Article  CAS  PubMed  Google Scholar 

  43. Zimmerman GA, McIntyre TM, Prescott SM, Stafforini DM: The platelet-activating factor signaling system and its regulators in syndromes of inflammation and thrombosis. Crit Care Med 2002, 30: S294-S301. 10.1097/00003246-200205001-00020

    Article  CAS  PubMed  Google Scholar 

  44. Sass JO, Ensenauer R, Roschinger W, Reich H, Steuerwald U, Schirrmacher O, Engel K, Haberle J, Andresen BS, Megarbane A, et al.: 2-Methylbutyryl-coenzyme a dehydrogenase deficiency: functional and molecular studies on a defect in isoleucine catabolism. Mol Genet Metab 2008, 93: 30–35.

    Article  CAS  PubMed  Google Scholar 

  45. Knebel LA, Zanatta A, Tonin AM, Grings M, Alvorcem Lde M, Wajner M, Leipnitz G: 2-Methylbutyrylglycine induces lipid oxidative damage and decreases the antioxidant defenses in rat brain. Brain Res 2012, 1478: 74–82.

    Article  CAS  PubMed  Google Scholar 

  46. Holden HM, Rayment I, Thoden JB: Structure and function of enzymes of the Leloir pathway for galactose metabolism. J Biol Chem 2003, 278: 43885–43888. 10.1074/jbc.R300025200

    Article  CAS  PubMed  Google Scholar 

  47. Novak EM, Lee EK, Innis SM, Keller BO: Identification of novel protein targets regulated by maternal dietary fatty acid composition in neonatal rat liver. J Proteomics 2009, 73: 41–49. 10.1016/j.jprot.2009.07.008

    Article  CAS  PubMed  Google Scholar 

  48. Boll M, Löwel M, Berndt J: Effect of unsaturated fatty acids on sterol biosynthesis in yeast. Biochimica et Biophysica Acta (BBA) - Lipids Lipid Metabol 1980, 620: 429–439. 10.1016/0005-2760(80)90134-4

    Article  CAS  Google Scholar 

  49. Galjart NJ, Morreau H, Willemsen R, Gillemans N, Bonten EJ, D’Azzo A: Human lysosomal protective protein has cathepsin a-like activity distinct from its protective function. J Biol Chem 1991, 266: 14754–14762.

    CAS  PubMed  Google Scholar 

  50. Wera S, Hemmings BA: Serine/threonine protein phosphatases. Biochem J 1995,311(Pt 1):17–29.

    CAS  PubMed Central  PubMed  Google Scholar 

  51. Foretz M, Carling D, Guichard C, Ferre P, Foufelle F: AMP-activated protein kinase inhibits the glucose-activated expression of fatty acid synthase gene in rat hepatocytes. J Biol Chem 1998, 273: 14767–14771. 10.1074/jbc.273.24.14767

    Article  CAS  PubMed  Google Scholar 

  52. Fu H, Subramanian RR, Masters SC: 14–3-3 proteins: structure, function, and regulation. Annu Rev Pharmacol Toxicol 2000, 40: 617–647. 10.1146/annurev.pharmtox.40.1.617

    Article  CAS  PubMed  Google Scholar 

  53. Mhawech P: 14–3–3 proteins–an update. Cell Res 2005, 15: 228–236. 10.1038/sj.cr.7290291

    Article  CAS  PubMed  Google Scholar 

  54. Koo SH, Flechner L, Qi L, Zhang X, Screaton RA, Jeffries S, Hedrick S, Xu W, Boussouar F, Brindle P, et al.: The CREB coactivator TORC2 is a key regulator of fasting glucose metabolism. Nature 2005, 437: 1109–1111. 10.1038/nature03967

    Article  CAS  PubMed  Google Scholar 

  55. Viollet B, Guigas B, Leclerc J, Hébrard S, Lantier L, Mounier R, Andreelli F, Foretz M: AMP-activated protein kinase in the regulation of hepatic energy metabolism: from physiology to therapeutic perspectives. Acta Physiologica 2009, 196: 81–98. 10.1111/j.1748-1716.2009.01970.x

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  56. Wada T, Kenmochi H, Miyashita Y, Sasaki M, Ojima M, Sasahara M, Koya D, Tsuneki H, Sasaoka T: Spironolactone improves glucose and lipid metabolism by ameliorating hepatic steatosis and inflammation and suppressing enhanced gluconeogenesis induced by high-fat and high-fructose diet. Endocrinology 2010, 151: 2040–2049. 10.1210/en.2009-0869

    Article  CAS  PubMed  Google Scholar 

  57. McClung JK, Jupe ER, Liu XT, Dell’Orco RT: Prohibitin: potential role in senescence, development, and tumor suppression. Exp Gerontol 1995, 30: 99–124. 10.1016/0531-5565(94)00069-7

    Article  CAS  PubMed  Google Scholar 

  58. Mishra S, Murphy LC, Nyomba BL, Murphy LJ: Prohibitin: a potential target for new therapeutics. Trends Mol Med 2005, 11: 192–197. 10.1016/j.molmed.2005.02.004

    Article  CAS  PubMed  Google Scholar 

  59. Vessal M, Mishra S, Moulik S, Murphy LJ: Prohibitin attenuates insulin-stimulated glucose and fatty acid oxidation in adipose tissue by inhibition of pyruvate carboxylase. FEBS J 2006, 273: 568–576. 10.1111/j.1742-4658.2005.05090.x

    Article  CAS  PubMed  Google Scholar 

  60. Burton GR, Nagarajan R, Peterson CA, McGehee RE Jr: Microarray analysis of differentiation-specific gene expression during 3T3-L1 adipogenesis. Gene 2004, 329: 167–185.

    Article  CAS  PubMed  Google Scholar 

  61. Artal-Sanz M, Tavernarakis N: Prohibitin couples diapause signalling to mitochondrial metabolism during ageing in C. elegans. Nature 2009, 461: 793–797. 10.1038/nature08466

    Article  CAS  PubMed  Google Scholar 

  62. Wu X, Zhang L, Gurley E, Studer E, Shang J, Wang T, Wang C, Yan M, Jiang Z, Hylemon PB, et al.: Prevention of free fatty acid-induced hepatic lipotoxicity by 18beta-glycyrrhetinic acid through lysosomal and mitochondrial pathways. Hepatology 2008, 47: 1905–1915. 10.1002/hep.22239

    Article  CAS  PubMed  Google Scholar 

  63. Cousin SP, Hugl SR, Wrede CE, Kajio H, Myers MG Jr, Rhodes CJ: Free fatty acid-induced inhibition of glucose and insulin-like growth factor I-induced deoxyribonucleic acid synthesis in the pancreatic beta-cell line INS-1. Endocrinology 2001, 142: 229–240. 10.1210/en.142.1.229

    CAS  PubMed  Google Scholar 

  64. Kuo C-F, Hsieh C-H, Lin W-Y: Proteomic response of LAP-activated RAW 264.7 macrophages to the anti-inflammatory property of fungal ergosterol. Food Chem 2011, 126: 207–212. 10.1016/j.foodchem.2010.10.101

    Article  CAS  Google Scholar 

  65. Ramirez-Zacarias JL, Castro-Munozledo F, Kuri-Harcuch W: Quantitation of adipose conversion and triglycerides by staining intracytoplasmic lipids with Oil red O. Histochemistry 1992, 97: 493–497. 10.1007/BF00316069

    Article  CAS  PubMed  Google Scholar 

  66. Amacher DE, Martin BA: Tetracycline-induced steatosis in primary canine hepatocyte cultures. Fundam Appl Toxicol 1997, 40: 256–263. 10.1006/faat.1997.2389

    Article  CAS  PubMed  Google Scholar 

  67. Heider JG, Boyett RL: The picomole determination of free and total cholesterol in cells in culture. J Lipid Res 1978, 19: 514–518.

    CAS  PubMed  Google Scholar 

  68. Candiano G, Bruschi M, Musante L, Santucci L, Ghiggeri GM, Carnemolla B, Orecchia P, Zardi L, Righetti PG: Blue silver: a very sensitive colloidal Coomassie G-250 staining for proteome analysis. Electrophoresis 2004, 25: 1327–1333. 10.1002/elps.200305844

    Article  CAS  PubMed  Google Scholar 

  69. Li Q, Huang J, Liu S, Li J, Yang X, Liu Y, Liu Z: Proteomic analysis of young leaves at three developmental stages in an albino tea cultivar. Proteome Sci 2011, 9: 44. 10.1186/1477-5956-9-44

    Article  PubMed Central  PubMed  Google Scholar 

Download references

Acknowledgements

The work was supported by the Key Project of National Technology Innovation System for Tea Industry in the People’s Republic of China (CARS-23), the natural science foundation of Hunan Province (13JJ4067), National Science and Technology Support Program (2012bad33b11), National Natural Science Foundation of China (31100502) and Specialized Research Foundation for the Doctorial Program of Higher Education of China (20114320120004).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Guoan Luo.

Additional information

Competing interests

The authors declare that they have no competing interest.

Authors’ contributions

ZHL conceived of the study, and participated in all the experiment. QL participated in the 2-DE experiments, mass spectrometry analysis and western blotting analysis. JAH participated in participated in the design of the study. QLL participated in the cell experiments. YJY participated in the 2-DE experiments. HYL participated in western blotting analysis. WJX participated in the critical review of the manuscript. YL participated in the protein identification. SZ participated in the protein identification. BT participated in cell experiments. GAL conceived of the study, and participated in its design and coordination. All authors read and approved the final manuscript.

Zhonghua Liu, Qin Li contributed equally to this work.

Authors’ original submitted files for images

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Liu, Z., Li, Q., Huang, J. et al. Proteomic analysis of the inhibitory effect of epigallocatechin gallate on lipid accumulation in human HepG2 cells. Proteome Sci 11, 32 (2013). https://doi.org/10.1186/1477-5956-11-32

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1477-5956-11-32

Keywords