Transcriptomic and proteomic analysis of human hepatic stellate cells treated with natural taurine

  • Authors:
    • Jian Liang
    • Xin Deng
    • Fa-Sheng Wu
    • Yan-Fang Tang
  • View Affiliations

  • Published online on: March 20, 2013     https://doi.org/10.3892/mmr.2013.1389
  • Pages: 1442-1452
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The aim of this study was to investigate the differential expression of genes and proteins between natural taurine (NTau)‑treated hepatic stellate cells (HSCs) and control cells as well as the underlying mechanism of NTau in inhibiting hepatic fibrosis. A microculture tetrazolium (MTT) assay was used to analyze the proliferation of NTau‑treated HSCs. Flow cytometry was performed to compare the apoptosis rate between NTau-treated and non‑treated HSCs. Proteomic analysis using a combination of 2-dimensional gel electrophoresis (2DE) and mass spectrometry (MS) was conducted to identify the differentially expressed proteins. Microarray analysis was performed to investigate the differential expression of genes and real-time polymerase chain reaction (PCR) was used to validate the results. The experimental findings obtained demonstrated that NTau decreased HSC proliferation, resulting in an increased number of cells in the G0/G1 phase and a reduced number of cells in the S phase. Flow cytometric analysis showed that NTau-treated HSCs had a significantly increased rate of apoptosis when compared with the non‑treated control group. A total of 15 differentially expressed proteins and 658 differentially expressed genes were identified by 2DE and MS, and microarray analysis, respectively. Gene ontology (GO) functional analysis indicated that these genes and proteins were enriched in the function clusters and pathways related to cell proliferation, cellular apoptosis and oxidation. The transcriptome and proteome analyses of NTau-treated HSCs demonstrated that NTau is able to significantly inhibit cell proliferation and promote cell apoptosis, highlighting its potential therapeutic benefits in the treatment of hepatic fibrosis.

Introduction

Hepatic fibrosis refers to the excessive accumulation of extracellular matrix (ECM) components in the liver. It occurs in most types of chronic liver diseases, including liver cirrhosis, liver failure, and portal hypertension, and often requires liver transplantation (1). The activation of hepatic stellate cells (HSCs) is an important event by which this cell type, which is otherwise quiescent, expresses α-smooth muscle actin (α-SMA), assumes a myofibroblastic phenotype and synthesizes fibrillar collagens (2,3). Therefore, the inhibition of HSC proliferation, the regulation of the HSC cell cycle, and the facilitation of HSC apoptosis are important therapeutic approaches for hepatic fibrosis-related liver diseases.

Taurine (2-aminoethanesulfonic acid) is an organic acid which is abundant in the human body. Natural taurine (NTau) has emerged as an alternative candidate for therapeutic intervention since it is effective in preventing hepatic fibrosis and reducing cirrhosis (4,5). Supplementation with exogenous taurine is able to extensively inhibit the deposition of ECM and mitigate the degree of hepatic fibrosis (2). Several studies have focused on the specific gene regulation associated with the protective effect of taurine against hepatic damage (68), but the genome-wide genes, proteins and functional pathways underlying the hepatic protection have yet to be fully elucidated.

Gene-expression profiling through microarray analysis may shed light on useful clues to the taurine-mediated gene regulation. Furthermore, a proteomics approach may also be used to elucidate global protein expression and facilitate the discovery of potential drug targets. However, studies using microarray or proteomic technologies to investigate the molecular mechanism of taurine treatment for liver diseases have not been previously been conducted. Therefore, an integrative analysis of transcriptome and proteome levels was designed to illuminate the changes of gene and protein expression in human HSCs treated with NTau.

Materials and methods

NTau extraction

NTau (2-aminoethanesulfonic acid) was extracted from black clams (Meretrix meretrix L.). Briefly, the clam meats were weighed and minced in an electrical blender (4000 rpm), for ~10 sec. The mince was further homogenized for 30 min after adding distilled water (1 liter). The mixture was boiled in water for 30 min, followed by filtering through 4 layers of gauze. The residue on the top of the gauze was discarded, and the filtrate was then centrifuged (3000 rpm) to obtain the supernatant, which was then de-acidified with HCl (HCl:H2O=3). After centrifuging, the proteins were adjusted to a pH of 10 with a NaOH (20%) aqueous solution to yield the de-alkalinated protein. Following adjustment of the pH value to 5, the supernatant was further condensed. The other unwanted amino acids and pigments were removed by column chromatography using strong-acid cation-exchange resin as the solid phase and eluting with distilled water. The resultant NTau was quantitatively measured by high-performance liquid chromatography (HPLC), and the purity of the NTau was determined to be 98.8%.

Cell culture

LX-2 human HSCs (purchased from the Cell Bank at Xiangya Central Experiment Laboratory of Central South University, Changsha, China) were cultivated at 37°C in Dulbecco’s modified Eagle’s medium (DMEM; Gibco-BRL, Carlsbad, CA, USA), and were supplemented with 10% fetal bovine serum (FBS) and penicillin-streptomycin in a 5% CO2 humidified incubator. Cells (1×104 or 5×105) were seeded in 96-well plates or 35-mm dishes. The cells were cultured in serum-free medium for 24 h and then treated with 40 mM NTau for 48 h.

Cell proliferation analysis

The proliferation activity of the LX-2 cells was measured by a microculture tetrazolium (MTT) colorimetric assay. LX-2 cells (1×104 cells/ml) were cultured in 96-well plates in DMEM with 10% FBS medium and then tranferred to serum-free medium for an additional 24 h, and triplicate wells of cells were incubated for 48 h in the presence 0–50 mM NTau. Cells in the various treatment groups were then incubated with MTT [5 mg/ml in phosphate-buffered solution (PBS)] for 4 h before harvesting. The optical density was measured using an ELISA reader at 570 nm with a reference wavelength of 630 nm.

Cell cycle analysis

The LX-2 cells were incubated for 24 h in a 5-ml cell culture flask in DMEM with 10% FBS medium and then cultured in serum-free medium for an additional 24 h. The cells were treated with 40 mM NTau for 48 h and then fixed with 70% ice-cold ethanol. The fixed cells were permeabilized with PBST (137 mM NaCl, 3 mM KCl, 8 mM NA2HPO4 and 0.1% Tween-20; pH 7.4) and then stained with 100 mg/l RNase A and 50 mg/l propidium iodide (PI) in the dark for cell cycle analysis. Cell cycle analysis was performed on a Coulter ELITE flow cytometer (BD Biosciences, Franklin Lakes, NJ, USA) through a 488-nm (LP) filter. The data were analyzed using the MultiCycle AV software (Phoenix Flow Systems, San Diego, CA, USA) for cell cycle distribution.

Apoptosis detection

LX-2 cells were incubated for 48 h in the presence 40 mM NTau and then re-suspended in 100 μl buffer containing calcium ions. The cells were treated with 5 μl Annexin V-FITC dye for 20–30 min and then with 5 μl PI dye for 5 min. The cell concentration was adjusted to ~1×105/ml by adding the appropriate amount of calcium ion-containing buffer. The cells were loaded on a flow cytometer (Coulter® Elite) within 1 h. Apoptotic analysis was performed using a Coulter® Elite flow cytometer (Beckman Coulter, Miami, FL, USA) with excitation and emission wavelengths of 488 and 530 nm, respectively.

Two-dimensional electrophoresis (2DE)

LX-2 cell lysates were collected and centrifuged at 14,000 rpm for 10 min at 4°C. The supernatants were analyzed by 2DE, and isoelectric focusing (IEF) was performed using an IPGphor IEF system (Bio-Rad, Hercules, CA, USA). The protein extract (200 μg) was mixed with rehydration buffer to 350 μl and loaded onto 17-cm, immobilized, nonlinear pH gradient (IPG) dry strips (pH 4–7; Bio-Rad). The IPG strips were equilibrated for 15 min in an equilibration buffer (6 M urea, 30% glycerol, 2% SDS and 50 mM Tris-HCl) containing 10 mg/ml dithiothreitol (DTT), followed by 15 min in an equilibration buffer containing 40 mg/ml iodoacetamide. Following equilibration, strips were applied to 12% sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE) gels and sealed with agarose sealing solution. Following electrophoresis, the SDS-PAGE gels were silver stained. Stained gels were scanned using an image scanner (Amersham Biosciences, Piscataway, NJ, USA) in transmission mode. Analysis of the gels was accomplished using the PDQuest analysis software (Bio-Rad) including background subtraction, spots detection and the establishment of a reference gel. Protein spots were selected based on the criterion of >2-fold variation of expression between NTau-treated and control samples.

Mass spectrometry (MS) analysis

MS was performed on a UPLC-ESI-MS/MS (Waters/Micromass, Manchester, UK) equipped with an electrospray ionization (ESI) source. Data-dependent analysis was employed (the 3 most abundant ions in each cycle): 1 sec MS m/z 350-1,600, and max 5 sec MS/MS m/z 50-2,000 (continuum mode), with 50 sec dynamic exclusion. The positive-ion mode was employed, and the capillary voltage was set at 3.0 kV. The cone voltage was set at 35 V to investigate the intensities and distribution of ions in the mass spectra of samples. The MS/MS spectra were processed, searched using ProteinLynx Global SERVER™ (PLGS) v2.3 (Waters/Micromass), and searched against the NCBInr database by MASCOT (http://www.matrixscience.co.uk) using the following constraints: only tryptic peptides with up to 2 missed cleavage sites were allowed and 0.3-Da mass tolerances for MS and MS/MS fragment ions. The results were filtered by a peptide score of ≥30.

Western blot analysis

Total proteins in LX-2 cell lysates were quantified by the Bradford method (9,10) and analyzed on 12% SDS-PAGE gels. The gels were transferred onto a nitrocellulose membrane using a Trans-Blot SD apparatus (Bio-Rad). The membrane was incubated with anti-IgY [dilution 1:1,000 in Tris-buffered saline (TBS)] followed by incubation with secondary antibody (dilution 1:5,000 in TBS). Visualization of the protein bands was achieved by the chemiluminescence method, and the films were developed and fixed. Glyeraldehyde-3-phosphate dehydrogenase (GAPDH) was used as an internal reference.

Gene ontology (GO) analysis

GO is a stratified tree structure for the analysis of the functions of genes and proteins (11). The 3 hierarchical principles of GO are ‘Biological Process’ (BP), ‘Cellular Component’ (CC) and ‘Molecular Function’ (MF). We analyzed the functional distribution of differential gene expression and protein production over the 3 principles. To accurately detect significantly over-represented GO terms, the Database for Annotation, Visualization, and Integrated Discovery (DAVID) tool (http://david.abcc.ncifcrf.gov) was used by analyzing into the fourth layers (12). The threshold value of group membership counts was set at 3, and the EASE score was set at 0.1. Then, the functional annotation clustering tool in DAVID was used to cluster functionally related annotations into groups for a 2D view of the related gene-term relationship (12). We ranked the importance of annotation groups with an enrichment score. In addition, we also used the DAVID tool to map differential gene expression and protein production into the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways database (13) to facilitate the biological interpretation in a network context. Fisher’s exact test was used and P<0.05 was considered to indicate a statistically significant difference.

Microarray analysis

A genome-wide 70-mer oligonucleotide microarray including 22,000 well-characterized human genes (14) was obtained from CapitalBio Corporation (Beijing, China) to investigate the expression profiling of NTau-treated LX-2 cells. The cDNA targets were prepared from 5 μg total RNA and were labeled with fluorescent dyes (Cy5 and Cy3-dCTP) by the Eberwine linear RNA amplification method and a subsequent enzymatic reaction (14). The RNA samples from the NTau-treated LX-2 cells were labeled with Cy3-dUTP and named as ‘1.’ The RNA samples from the blank control cells were labeled with Cy5-dUTP and named as ‘2.’ We then prepared the hybridization solution in hybridization buffer (25% formamide, 3X SSC, 0.2% SDS and 5X Denhardt’s solution) and hybridized it with sample at 42°C overnight in a humid environment. The hybridized microarrays were scanned with a confocal LuxScan™ scanner (CapitalBio Corporation, Beijing, China) at 2 wavelengths to detect emission from both Cy3 and Cy5. The images obtained were then analyzed using LuxScan™ 3.0 software (CapitalBio Corporation). Then, an intensity-dependent locally weighted scatterplot-smoothing regression (LOWESS) algorithm was used to normalize the 2-channel ratio values by an R package (15).

Gene set enrichment analysis (GSEA)

GSEA is a software (https://www.broad.harvard.edu/gsea/) for searching in predefined gene sets (ex. pathways) and ranking genes to identify significant biological changes in microarray data sets (16). First, ratios of filtered genes were operated by logarithm function to the base 1.5, and the input ‘rnk’ file was made based on the ranked value of log1.5(ratio). These expression matrices were exported to GSEA software and searched against the background dataset of ‘c4: computational gene sets’ with ‘GSEA Pre-ranked’ option. All the default settings except ‘gene set permutation’ with 1,000 iterations were used for the analysis.

RNA extraction and real-time polymerase chain reaction (PCR)

Total RNA was extracted using TRIzol (Invitrogen, Gaithersburg, MD, USA) reagent according to the manufacturer’s instructions. RNA was purified using the NucleoSpin RNA Clean-up kit (Macherey-Nagel, Düren, Germany). RNA quality from each sample was assessed by visualization of the 28S/18S ribosomal RNA ratio on 1% agarose gels. First-strand cDNA was synthesized using 1 μg total RNA in a 20-μl final volume by reverse transcription utilizing ReverTra Ace® reverse transcriptase (Toyobo, Co., Ltd., Osaka, Japan) with random hexamer primers or oligo(dT)18 primers (Invitrogen). PCR was performed using 0.5 μl cDNA, with specific primers and Ex Taq™ Polymerase (Takara Bio, Inc., Otsu, Japan) in a volume of 12.5 μl. The PCR products were then separated on 1.5% agarose gels. The real-time PCR reactions were performed using iQ™ SYBR®-Green Supermix kit according to the manufacturer’s instructions (Bio-Rad). RNA was amplified using the ABI Prism 7500 Sequence Detection system (Applied Biosystems, Carlsbad, CA, USA). The primers (Invitrogen) are shown in Table I. For all the real-time PCR experiments, negative controls were a non-reverse transcriptase reaction, and a non-sample reaction (data not shown). GAPDH was amplified as an internal standard.

Table I

Primers used for real-time PCR of the cox5a, cox6c, ndufb1, ndufc1 and tgfβ1|1 genes.

Table I

Primers used for real-time PCR of the cox5a, cox6c, ndufb1, ndufc1 and tgfβ1|1 genes.

GenesPrimerSequence (5′-3′)Temperature (°C)Product size (bp)
cox5aForward TAAACCGCATGGATGGGC49177
Reverse AGTTCAAACTCATTTCCCTTTTATT
cox6cForward GGGGTTGCAGCTTTGTAT349112
Reverse CAGCCTTCCTCATCTCCT
ndufc1Forward CCGAATGCCAAACCTGAC49127
Reverse ATTCCAGCCCATTTCTTC
ndufb1Forward TTCCCTGTTGCCCTTGGT53.1158
Reverse AGCCGTTCATCACTCTTTCTGT
tgfβ1|1Forward TTCTGCTGCGTCAGTTGC57.4154
Reverse TGAGCGCCGAGATGTAGTT
gapdhForward GACCTGACCTGCCGTCTA56148
Reverse AGGAGTGGGTGTCGCTGT
Statistical analysis

All experiments were performed ≥3 times with triplicate measurements, and data are expressed as the mean ± standard error of the mean (SEM). Statistical analysis was performed using the R software (version 2.9.2, http://www.r-project.org/). Student’s t-test was performed to evaluate the differences of cell proliferation rate among the groups treated with different concentrations of NTau. The Student’s t-test was also used to evaluate the gene expression changes in real-time PCR results between 2 phenotypes. The Chi-square test was used to assess the effect of NTau on the cell cycle of HSCs. Fisher’s exact test was used to identify the significant GO terms of target genes relative to genome backgrounds. P<0.05 was considered to indicate a statistically significant difference.

Results

Effect of NTau on cell proliferation and cell cycle of HSCs

As shown in Fig. 1, an inverse correlation between the proliferation of LX-2 cells and the concentration of NTau was detected. Treatment with different concentrations of NTau significantly inhibited the growth of LX-2 cells when compared with that of the non-treated control cells (one-way ANOVA, P<0.005). The growth rate of LX-2 cells in the presence of 30–40 mmol/l NTau was reduced by 43% compared with that of the non-treated cells. As assessed by cell cycle analysis, the percentage of LX-2 cells in the G0/G1 phase increased from 43.9 to 50.9% in response to 40 mmol/l NTau, while exposure of the LX-2 cells to 40 mmol/l NTau for 48 h caused a 1- to 3-fold reduction (P<0.04) of the S- and G2/M-phase cell populations compared with those in the non-treated control cells.

Detection of NTau-induced apoptosis in HSCs

To investigate the mechanism underlying the growth-inhibitory effects of NTau on HSCs, the cell-apoptosis analysis of NTau-treated LX-2 cells was performed. As shown in Fig. 2, the increase in apoptotic cells in the NTau-treated LX-2 cells was comparable to that in the non-treated control cells (13.6±3.3 vs. 4.65±1.1%, P<0.05). These findings suggest that the induction of cellular apoptosis contributed, at least in part, to the HSC growth-inhibiting effects of NTau.

Identification of differentially expressed proteins in HSCs following NTau treatment

The protein profiles of HSCs were analyzed by 2DE and visualized using the PDQuest image analysis software. By comparing the protein profiles of the NTau-treated and non-treated control HSCs, 15 differentially expressed proteins were successfully identified. We picked up protein spots in the 2D gel image with identified changes and prepared them for MS analysis. Table II summarizes the code name, relative molecular weight, isoelectric point, and peptide fragment coverage for the differentially expressed proteins. The most significantly upregulated expression was of CAA32649, MYL9, PSMB6, ANXA1, MDH1, HSPB1, LASP1, LOC100134370, and SOD1, while ATP5H, BAF82933, ECHS1, PRDX2, HNRNPA2B1, and BAG36698 showed the most markedly downregulated protein expression.

Table II

Differentially expressed proteins in taurine-treated vs. control HSCs.

Table II

Differentially expressed proteins in taurine-treated vs. control HSCs.

Protein_IDGIGeneLog2 (ratio)DescriptionRelative molecular weight (kDa)Isoelectric pointPeptide fragment coverage (%)
Upregulated
 Protein 139428317CAA326494.247Unnamed protein product59.55.1727
 Protein 143129568111MYL93.295Myosin regulatory light chain 9 isoform A19.84.8062
 Protein 1380558528PSMB62.621Proteasome (prosome, macropain) subunit, β type, 625.34.8026
 Protein 64502101ANXA11.483Annexin I38.76.5753
 Protein 85174539MDH11.180Cytosolic malate dehydrogenase36.46.9117
 Protein 34504517HSPB10.989Heat shock protein β-122.85.9849
 Protein 75453710LASP10.953LIM and SH3 protein 129.76.6134
 Protein 1169204721LOC1001343700.703Predicted: hypothetical protein54.26.3243
 Protein 52982080SOD10.504Superoxide dismutase 1, soluble15.95.8745
Downregulated
 Protein 105453559ATP5H−0.663ATP synthase, H+ transporting, mitochondrial F0 complex, subunit d isoform A18.55.2147
 Protein 2158261511BAF82933−0.704Unnamed protein product49.54.864
 Protein 4194097323ECHS1−0.825Mitochondrial short-chain enoyl-coenzyme A hydratase 1 precursor31.48.3430
 Protein 99955007PRDX2−1.135Peroxiredoxin 221.85.4438
 Protein 95314043072HNRNPA2B1−1.864Heterogeneous nuclear ribonucleoprotein A2/B1 isoform B137.48.9715
 Protein 1165189054178BAG36698−2.812Unnamed protein product66.07.6217

[i] HSCs, hepatic stellate cells.

Functional analysis of differentially expressed proteins in HSCs following NTau treatment and validation of proteomic data

We analyzed the functional enrichment of the differentially expressed proteins using the DAVID tool based on their annotation keywords from the UniProt database (17). Significant protein functions terms and corresponding proteins were identified (Fisher’s exact test, P<0.05). As shown in Table III, proteins that corresponded to the category of ‘acetylation,’ ‘direct protein sequencing,’ ‘antioxidant,’ ‘cytoplasm’ and ‘oxidoreductase’ were significantly affected in the NTau-treated HSCs.

Table III

Functional enrichment analysis based on the annotation keywords of proteins.

Table III

Functional enrichment analysis based on the annotation keywords of proteins.

TermP-valueProteinsFold enrichment
Acetylation6.8E-09PRDX2, ATP5H, MDH1, MYL9, HSPB1, LASP1, SOD1, ECHS120.79
Direct protein sequencing1.0E-06PRDX2, PSMB6, ATP5H, MDH1, HSPB1, HNRNPA2B1, SOD1, ECHS110.04
Antioxidant5.5E-03PRDX2, SOD1328.10..
Cytoplasm5.9E-03PRDX2, PSMB6, MDH1, LASP1, SOD1, ANXA14.08
Oxidoreductase4.7E-02PRDX2, MDH1, SOD17.70

[i] Fisher’s exact test, P<0.05

Among the differentially expressed proteins identified successfully by MS, the upregulated proteins ANXA1 and PSMB6 and the downregulated proteins ECHS1 and PRDX2 were selected and subjected to western blot analysis. As shown in Fig. 3, NTau treatment significantly upregulated the expression of ANXA1 and downregulated the expression of ECHS1 and PRDX2 in HSCs. The expression of PSMB6 was not significantly different following NTau treatment. Therefore, western blot analysis of the differentially expressed proteins confirmed the reliability and validity of the proteomic high throughput experiments.

Differential gene expression in HSCs following NTau treatment

According to the filtering principles described above, 6,109 normally expressed genes (28.38%) with high confidence were screened among 21,522 genes. By applying the threshold of 1.5-fold change for an intensity ratio of NTau-treated vs. control HSCs, 658 genes (3.06%) were shown to be differentially expressed in NTau-treated HSCs. Among the differentially expressed genes, 241 were upregulated (1.12%) and 417 were downregulated (1.94%). The top 9 upregulated and the top 10 downregulated genes are presented in Table IV.

Table IV

Top 9 upregulated and top 10 downregulated genes in taurine-treated vs. control HSCs.

Table IV

Top 9 upregulated and top 10 downregulated genes in taurine-treated vs. control HSCs.

GeneRefSeq_IDDescription log1.5(ratio)
Top 9 upregulated
hdac3NM_003883Histone deacetylase 31.8950
tgfβ1i1NM_015927Transforming growth factor β1-induced transcript 11.8773
tmem120aNM_031925Transmembrane protein induced by tumor necrosis factor-α1.8526
cyp2e1NM_000773Cytochrome P450, family 2, subfamily E, polypeptide 11.7893
hrh1NM_000861Histamine receptor H11.7789
seltNM_016275Selenoprotein T1.6962
ccdc86NM_024098Coiled-coil domain containing 861.6648
mif4gdNM_020679MIF4G domain containing1.6570
psapNM_002778Prosaposin (variant Gaucher disease and variant metachromatic leukodystrophy)1.6430
Top 10 downregulated
nucb2NM_005013Nucleobindin 2−2.7135
adam9NM_003816A disintegrin and metalloproteinase domain 9 (meltrin γ)−2.7336
tpp2NM_003291Tripeptidyl peptidase II−2.7350
abce1NM_002940ATP-binding cassette, sub-family E (OABP), member 1−2.7956
mtif2NM_002453Mitochondrial translational initiation factor 2−2.8257
pkn2NM_006256Protein kinase N2−3.2762
ndufb1NM_004545NADH dehydrogenase (ubiquinone) 1 β subcomplex, 1, 7 kDa−3.6023
brcc3NM_024332 BRCA1/BRCA2-containing complex, subunit 3−3.6603
adamtsl3NM_207517ADAMTS-like 3−3.7298
adam9NM_003816A disintegrin and metalloproteinase domain 9 (meltrin γ)−5.9415

[i] HSCs, hepatic stellate cells.

Functional categorization and clustering for the differentially expressed genes

The differentially expressed genes were then categorized according to their GO function using the DAVID tool. Six function categories of ‘Molecular Function’ (MF), 51 function categories of ‘Cellular Component’ (CC), and 42 function categories of ‘Biological Process’ (BP) were significantly enriched in the differentially expressed genes (Fisher’s exact test, P<0.05). Fig. 4 lists the top 5 significantly enriched GO terms identified after screening with a threshold of false discovery rate (FDR) of <0.01. Functional categorization of the GO terms demonstrated that these differentially expressed genes were strongly associated with biological processes of ‘NADH reduction and oxidation reaction’ and ‘RNA processing’. Based on the analysis above, the heuristic fuzzy clustering was used to classify the groups of similar annotations by the κ statistic values (18). Table V provides a visualized network of the 9 significantly enriched GO terms (geometric mean of member’s P-value, P<0.05). Clusters 1, 2, 3, 4, 7 and 9 correspond to the categories of ‘cellular transport and translation,’ ‘oxidant reaction’ and ‘mitosis process,’ respectively. This suggests a crucial role of these biological processes in NTau-treated HSCs.

Table V

Functional annotation clustering analysis of the differentially expressed genes.

Table V

Functional annotation clustering analysis of the differentially expressed genes.

CategoryTermPercentageP-valueFold enrichment
Functional group 1Geo: 4.6040E-15
 GOTERM_CC_4 GO:0044424-intracellular part0.68734.45E-221.2591
 GOTERM_CC_4 GO:0043229-intracellular organelle0.59377.00E-171.2999
 GOTERM_CC_4 GO:0043231-intracellular membrane-bound organelle0.53495.09E-161.3499
 GOTERM_CC_4 GO:0005634-nucleus0.32542.83E-051.2661
Functional group 2Geo: 1.4710E-9
 GOTERM_CC_4 GO:0019866-organelle inner membrane0.06032.33E-113.6125
 GOTERM_CC_4 GO:0031967-organelle envelope0.08252.28E-102.6796
 GOTERM_CC_4 GO:0044429-mitochondrial part0.07786.55E-102.6988
 GOTERM_CC_4 GO:0005743-mitochondrial inner membrane0.05409.73E-103.4607
 GOTERM_CC_4 GO:0005739-mitochondrion0.11272.203E-092.1238
 GOTERM_CC_4 GO:0044455-mitochondrial membrane part0.03332.326E-095.26001
 GOTERM_CC_4 GO:0031966-mitochondrial membrane0.06034.029E-093.0154
 GOTERM_CC_4 GO:0005740-mitochondrial envelope0.06194.501E-092.9486
 GOTERM_CC_4 GO:0031090-organelle membrane0.14295.406E-091.8718
 GOTERM_CC_4 GO:0005746-mitochondrial respiratory chain0.02542.795E-086.2282
Functional group 3Geo: 5.5926E-7
 GOTERM_CC_4 GO:0005743-mitochondrial inner membrane0.05409.73E-103.4607
 GOTERM_CC_4 GO:0044455-mitochondrial membrane part0.03332.33E-095.2601
 GOTERM_CC_4 GO:0005746-mitochondrial respiratory chain0.02542.80E-086.2282
 GOTERM_CC_4GO:0030964-NADH dehydrogenase complex (quinone)0.01751.56E-067.3688
 GOTERM_CC_4 GO:0005747-mitochondrial respiratory chain complex I0.01751.56E-067.3688
 GOTERM_CC_4 GO:0045271-respiratory chain complex I0.01751.56E-067.3688
 GOTERM_MF_4GO:0003954-NADH dehydrogenase activity0.01901.56E-066.5705
 GOTERM_MF_4 GO:0016655-oxidoreductase activity, acting on NADH or NADPH, quinone or similar compound as acceptor0.01904.22E-065.9640
 GOTERM_BP_4 GO:0006120-mitochondrial electron transport, NADH to ubiquinone0.01592.38E-056.2590
 GOTERM_BP_4GO:0042773-ATP synthesis coupled electron transport0.01758.01E-054.8297
Functional group 4Geo: 3.9490E-6
 GOTERM_CC_4GO:0044428-nuclear part0.11751.01E-082.0090
 GOTERM_CC_4 GO:0044451-nucleoplasm part0.05401.50E-052.2883
 GOTERM_CC_4 GO:0005654-nucleoplasm0.05872.47E-052.1445
 GOTERM_CC_4GO:0031981-nuclear lumen0.06986.50E-051.9002
Functional group 5Geo: 1.9522E-4
 GOTERM_BP_4 GO:0046907-intracellular transport0.07149.11E-051.8540
 GOTERM_BP_4GO:0015031-protein transport0.06980.00011.8571
 GOTERM_BP_4 GO:0045184-establishment of protein localization0.07140.00021.7769
 GOTERM_BP_4 GO:0006886-intracellular protein transport0.04600.00032.1064
 GOTERM_BP_4 GO:0051649-establishment of cellular localization0.07940.00041.6695
Functional group 6Geo: 0.0010
 GOTERM_BP_4 GO:0051246-regulation of protein metabolic process0.03810.00032.2997
 GOTERM_BP_4 GO:0009889-regulation of biosynthetic process0.02860.00072.5580
 GOTERM_MF_4 GO:0003743-translation initiation factor activity0.01590.00074.0893
 GOTERM_BP_4 GO:0006446-regulation of translational initiation0.01270.00085.1161
 GOTERM_BP_4 GO:0006417-regulation of translation0.02540.00092.7050
 GOTERM_BP_4 GO:0022618-protein-RNA complex assembly0.01900.00193.0432
 GOTERM_BP_4 GO:0031326-regulation of cellular biosynthetic process0.02540.00202.4904
 GOTERM_BP_4 GO:0006412-translation0.05710.00281.6837
Functional group 7Geo: 0.0073
 GOTERM_CC_4 GO:0012505-endomembrane system0.08250.00151.5603
 GOTERM_CC_4 GO:0005783-endoplasmic reticulum0.07300.00181.6022
 GOTERM_CC_4 GO:0044432-endoplasmic reticulum part0.04440.01311.6327
 GOTERM_CC_4GO:0042175-nuclear envelope-endoplasmic reticulum network0.03970.02081.6219
 GOTERM_CC_4 GO:0005789-endoplasmic reticulum membrane0.03810.02911.5893
Functional group 8Geo: 0.0225
 GOTERM_BP_4 GO:0007067-mitosis0.02540.01062.0735
 GOTERM_BP_4GO:0000087-M phase of mitotic cell cycle0.02540.01152.0554
 GOTERM_BP_4GO:0000279-M phase0.02700.03591.7425
 GOTERM_BP_4GO:0022403-cell cycle phase0.03020.05881.5700
Functional group 9Geo: 0.0346
 GOTERM_CC_4GO:0005635-nuclear envelope0.02060.02472.0575
 GOTERM_CC_4GO:0005637-nuclear inner membrane0.00630.02656.0643
 GOTERM_CC_4GO:0044453-nuclear membrane part0.01270.04672.4257
 GOTERM_CC_4GO:0031965-nuclear membrane0.01590.04692.1180

[i] GO, gene ontology; CC, cellular component; MF, molecular function; BP, biological process.

Pathway-based (GSEA) microarray analysis of NTau-treated HSCs and validation of microarray analysis

mRNA expression profiling using GSEA microarrays and quantitative PCR (qPCR) was performed in NTau-treated HSCs. Analysis of the expression of individual mRNAs demonstrated 2 different patterns of expression. A number of genes including nucb2, adam9, tpp2, mtif2, abce1, pkn2, ndufb1, brcc3, adamtsl3 and gbp1 correlated with reduction and oxidation of NADH showed a reduced expression in the NTau-treated HSCs. The second group, consisting of genes related to cell proliferation and cell cycle regulation including hdac3, tgfβ1i1 and hrh1, showed an increased expression in NTau-treated HSCs. We selected 2 of the gene sets ‘Module 62’ and ‘MORF_RAD21’ to illustrate the enrichment scores as shown in Fig. 5, which was driven by the group of genes within a gene set that showed the highest correlation with NTau treatment.

Five selected candidate genes, snrpe, hnrph3, eif1ay, nucb2 and vim, were used as reference genes were assayed by qPCR in order to confirm the expression profiles found using microarray analysis (Fig. 6). Another 5 selected genes, cox5a, cox6c, ndufb1, ndufc1 and tgfβ1i1, were assayed to identify the difference between NTau-treated and control HSCs. The primers designed for real-time PCR are listed in Table I. As shown in Fig. 7, tgfβ1i1 mRNA showed a significantly increased expression level (2.26±0.41 vs. 1, P=0.01), which coincided with pathway-based (GSEA) microarray analysis progression in the regulation of cell proliferation group. However, cox5a mRNA (0.62±0.03 vs. 1, P=2×10−4), cox6c mRNA(0.51±0.07 vs. 1, P=1.3×10−3), ndufb1 mRNA(0.53±0.07 vs. 1, P=2.7×10−5), and ndufc1 mRNA (0.45±0.06 vs. 1, P=1.1×10−3) showed a reduced expression in the NTau-treated HSCs, which are known to play a role in the regulation of NADH dehydrogenase (ubiquinone) activity, which is involved in NADH oxidation.

Discussion

Taurine is a sulfur-containing β-amino acid with several potential therapeutic applications 19,20). Substantial progress has been made over the last 10–20 years in elucidating the bio-physiological function of taurine in the treatment of liver fibrosis. It has been reported that taurine is able to protect hepatocytes from chemically induced injury and mitigate the fibrosis of the liver (6,7,21). Although recent findings suggest that taurine is able to inhibit the proliferation of HSCs and cause a G0/G1 phase arrest (2), the exact mechanism has not yet been fully elucidated. During chronic liver injury, HSCs undergo a phenotypic transformation with the acquisition of myofibroblast-like features by increased proliferation and synthesis of ECM components and play a pivotal role in the formation of fibrosis (22). Therefore, the inhibition of HSC proliferation, the regulation of the cell cycle of HSCs, and the facilitation of HSC apoptosis are important therapeutic approaches for hepatic fibrosis-related liver diseases. The present study is congruent with the study by Chen (21), according to which taurine not only inhibits the proliferation of HSCs, but it is also able to promote HSC apoptosis.

The process of apoptosis is controlled by a diverse range of cell signals, which may originate either extracellularly or intracellularly. A cell initiates intracellular apoptotic signaling in response to stress, which may cause cell suicide. The binding of nuclear receptors by glucocorticoids, heat, radiation increase and intracellular calcium concentration, for example, by causing damage to the membrane, are all able to trigger the release of intracellular apoptotic signals by a damaged cell (23,24).

The proteins ANXA1, ECHS1 and PRDX2 were selected for validation by western blot analysis. Functional analysis showed that these proteins were related to the biological processes of ‘cellular apoptosis’ and ‘oxidation reaction.’ Since flow cytometric analysis has shown that taurine-treated HSCs had a significantly increased apoptosis rate compared to the control group, the proteomic analysis may reveal the relevant mechanism. Upregulated protein Annexin I belongs to a family of Ca2+-dependent phospholipid-binding proteins, which are able to change the intracellular calcium concentration and cause apoptosis. Reactive oxygen species (ROS) are closely associated with apoptotic induction (25) and downregulate PRDX2 protein, a kind of antioxidant enzyme. Since altered cellular oxidation-reduction one of the key events in apoptosis that affects the mitochondria (26), it may be involved in oxidation reaction and HSC apoptosis.

GO analysis indicated that reduction and oxidation of NADH had significant functional enrichment. Fuzzy heuristic clustering of GO categories suggested that intracellular components of HSCs were influenced by NTau treatment. Furthermore, many differentially expressed genes were classified into function clusters relating to reduction-oxidation of mitochondrial NADH and mitosis. A variety of key events in apoptosis focus on the mitochondria, including the release of caspase activators, such as cytochrome c, changes in electron transport and altered cellular oxidation-reduction (26,27).

Among the validated genes, upregulated tgfβ1i1, which is transforming growth factor β1-induced transcript 1, is involved in the negative regulation of cell proliferation (28). This means that the higher its expression level, the slower the cell proliferation. Its upregulated level may indicate taurine’s function in inhibiting the proliferation of LX-2 cells. The downregulated genes cox6c, cox5a, ndufb1, and ndufc1 are components of the electron transport chain in the mitochondrion. Therefore, we hypothesize that NTau may regulate the reduction-oxidation of NADH and thereby lead to the inhibition of HSC proliferation.

The pathogenesis of hepatic fibrosis involves the activation of HSCs. This procedure is accelerated by HSC proliferation and the progression of the cell cycle (29). NTau has been demonstrated to inhibit HSC proliferation and prevent HSCs in the G0/G1 phase from entering the S and G2/M phases by flow cytometric analysis, which suggests that NTau is able to modulate hepatic fibrosis. Functional clustering and GSEA analyses also showed that mitosis, and especially the M phase of mitotic cell cycle, was regulated by NTau treatment. Therefore, it was not only demonstrated that NTau was beneficial to hepatic fibrosis therapy, but valuable evidence to elucidate the underlying molecular mechanism by investigating responded genes and proteins was also provided.

It is worth noting that reduction-oxidation of NADH may play an important role in the protective effect of NTau against hepatic fibrosis, since GO functional clustering and GSEA analysis consistently came to the same results. The reduction-oxidation state is often used to describe the balance of NAD+/NADH and NADP+/NADPH in a biological system such as a cell (30). Oxidative stress is important in the pathogenesis of hepatic fibrosis, which is the result of deposition of excess ECM proteins produced by activated HSCs (6). NTau may be able to attenuate ROS production by modulating the balance of reduction-oxidation of NADH. Furthermore, the relationship between the reduction-oxidation of NADH and HSC proliferation or cell cycle regulation has been previously investigated. Zou et al(31) have demonstrated that ROS derived from NAD(P)H oxidases activated the phosphotidylinositol-3-kinase (PI3K)/Akt pathway, thus promoting cellular proliferation in HSCs.

Taurine may be synthesized chemically or extracted from natural sources. However, NTau is superior to synthetic taurine in promoting HSC apoptosis (32). The advantage of the present study is reflected in the selection of NTau for investigation of its mechanism in regulating HSC apoptosis. To the best of our knowledge, the present study provided for the first time evidence concerning taurine-mediated transcriptional changes in HSCs by microarray analysis. Additionally, proteomic approaches were used to delineate protein expression changes in NTau-treated HSCs. These variations correspond to biological processes such as ‘oxidant reaction’ and ‘mitosis process,’ which promote HSC apoptosis. While these observations systematically investigated the underlying mechanism of NTau in inhibiting the activation of HSCs, our data provide strong support for the use of NTau as a potential therapy for hepatic fibrosis.

Acknowledgements

This study was supported by the National Natural Science Foundation of China (81160433) and the Guangxi Natural Science Foundation (2010GXNSFA013217). The authors thank Tiandiyang Biology Corporation and the SysBiomics Bioinformatics Co., Ltd. (Beijing, China), for their instructive help with data analysis.

References

1 

Bataller R and Brenner DA: Liver fibrosis. J Clin Invest. 115:209–218. 2005. View Article : Google Scholar

2 

Chen YX, Zhang XR, Xie WF and Li S: Effects of taurine on proliferation and apoptosis of hepatic stellate cells in vitro. Hepatobiliary Pancreat Dis Int. 3:106–109. 2004.PubMed/NCBI

3 

Wu J and Zern MA: Hepatic stellate cells: a target for the treatment of liver fibrosis. J Gastroenterol. 35:665–672. 2000. View Article : Google Scholar : PubMed/NCBI

4 

Kerai MD, Waterfield CJ, Kenyon SH, Asker DS and Timbrell JA: Taurine: protective properties against ethanol-induced hepatic steatosis and lipid peroxidation during chronic ethanol consumption in rats. Amino Acids. 15:53–76. 1998. View Article : Google Scholar

5 

Balkan J, Dogru-Abbasoglu S, Kanbagli O, Cevikbas U, Aykac-Toker G and Uysal M: Taurine has a protective effect against thioacetamide-induced liver cirrhosis by decreasing oxidative stress. Hum Exp Toxicol. 20:251–254. 2001. View Article : Google Scholar : PubMed/NCBI

6 

Refik Mas M, Comert B, Oncu K, Vural SA, Akay C, Tasci I, Ozkomur E, Serdar M, Mas N, Alcigir G and Yener N: The effect of taurine treatment on oxidative stress in experimental liver fibrosis. Hepatol Res. 28:207–215. 2004.PubMed/NCBI

7 

Ghandforoush-Sattari M and Mashayekhi S: Evaluation of taurine as a biomarker of liver damage in paracetamol poisoning. Eur J Pharmacol. 581:171–176. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Miyazaki T, Karube M, Matsuzaki Y, Ikegami T, Doy M, Tanaka N and Bouscarel B: Taurine inhibits oxidative damage and prevents fibrosis in carbon tetrachloride-induced hepatic fibrosis. J Hepatol. 43:117–125. 2005. View Article : Google Scholar : PubMed/NCBI

9 

Bradford MM: A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem. 72:248–254. 1976. View Article : Google Scholar : PubMed/NCBI

10 

Stoscheck CM: Quantitation of protein. Methods Enzymol. 182:50–68. 1990. View Article : Google Scholar

11 

Ashburner M, Ball CA, Blake JA, et al: Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat Genet. 25:25–29. 2000. View Article : Google Scholar : PubMed/NCBI

12 

Huang da W, Sherman BT and Lempicki RA: Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 4:44–57. 2009.PubMed/NCBI

13 

Kanehisa M, Araki M, Goto S, Hattori M, Hirakawa M, Itoh M, Katayama T, Kawashima S, Okuda S, Tokimatsu T and Yamanishi Y: KEGG for linking genomes to life and the environment. Nucleic Acids Res. 36:D480–D484. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Patterson TA, Lobenhofer EK, Fulmer-Smentek SB, et al: Performance comparison of one-color and two-color platforms within the MicroArray Quality Control (MAQC) project. Nat Biotechnol. 24:1140–1150. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Yang YH, Dudoit S, Luu P, Lin DM, Peng V, Ngai J and Speed TP: Normalization for cDNA microarray data: a robust composite method addressing single and multiple slide systematic variation. Nucleic Acids Res. 30:e152002. View Article : Google Scholar : PubMed/NCBI

16 

Subramanian A, Tamayo P, Mootha VK, et al: Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 102:15545–15550. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Apweiler R, Bairoch A, Wu CH, et al: UniProt: the Universal Protein knowledgebase. Nucleic Acids Res. 32:D115–D119. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Huang da W, Sherman BT, Tan Q, Collins JR, Alvord WG, Roayaei J, Stephens R, Baseler MW, Lane HC and Lempicki RA: The DAVID Gene Functional Classification Tool: a novel biological module-centric algorithm to functionally analyze large gene lists. Genome Biol. 8:R1832007.PubMed/NCBI

19 

Kendler BS: Taurine: an overview of its role in preventive medicine. Prev Med. 18:79–100. 1989. View Article : Google Scholar : PubMed/NCBI

20 

Lourenço R and Camilo ME: Taurine: a conditionally essential amino acid in humans? An overview in health and disease. Nutr Hosp. 17:262–270. 2002.PubMed/NCBI

21 

Chen YX: Protective action of taurine on ischemia-reperfusion liver injury in rats and its mechanism. Zhonghua Yi Xue Za Zhi. 73:276–279. 318–379. 1993.(In Chinese).

22 

Friedman SL: Seminars in medicine of the Beth Israel Hospital, Boston. The cellular basis of hepatic fibrosis Mechanisms and treatment strategies. N Engl J Med. 328:1828–1835. 1993. View Article : Google Scholar

23 

Cotran RS, Kumar V and Collins T: Robbins Pathologic Basis of Disease. 6th edition. WB Saunders Co; Philadelphia: 1998

24 

Mattson MP and Chan SL: Calcium orchestrates apoptosis. Nat Cell Biol. 5:1041–1043. 2003. View Article : Google Scholar : PubMed/NCBI

25 

Simon HU, Haj-Yehia A and Levi-Schaffer F: Role of reactive oxygen species (ROS) in apoptosis induction. Apoptosis. 5:415–418. 2000. View Article : Google Scholar : PubMed/NCBI

26 

Green DR and Reed JC: Mitochondria and apoptosis. Science. 281:1309–1312. 1998. View Article : Google Scholar : PubMed/NCBI

27 

Camilleri-Broët S, Vanderwerff H, Caldwell E and Hockenbery D: Distinct alterations in mitochondrial mass and function characterize different models of apoptosis. Exp Cell Res. 239:277–292. 1998.PubMed/NCBI

28 

Shibanuma M, Mashimo J, Kuroki T and Nose K: Characterization of the TGF beta 1-inducible hic-5 gene that encodes a putative novel zinc finger protein and its possible involvement in cellular senescence. J Biol Chem. 269:26767–26774. 1994.PubMed/NCBI

29 

Wells RG: The role of matrix stiffness in hepatic stellate cell activation and liver fibrosis. J Clin Gastroenterol. 39:S158–S161. 2005. View Article : Google Scholar : PubMed/NCBI

30 

Ying W: NAD+/NADH and NADP+/NADPH in cellular functions and cell death: regulation and biological consequences. Antioxid Redox Signal. 10:179–206. 2008.

31 

Zou CG, Gao SY, Zhao YS, Li SD, Cao XZ, Zhang Y and Zhang KQ: Homocysteine enhances cell proliferation in hepatic myofibroblastic stellate cells. J Mol Med (Berl). 87:75–84. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Liang J, Deng X, Yang GY, Huang RB and Pang YS: Effect of natural taurine on serology and histology of hepatic fibrosis in rat. Journal of Guangxi Traditional Chinese Medical University. 9:3–5. 2006.(In Chinese).

Related Articles

Journal Cover

May 2013
Volume 7 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liang J, Deng X, Wu F and Tang Y: Transcriptomic and proteomic analysis of human hepatic stellate cells treated with natural taurine. Mol Med Rep 7: 1442-1452, 2013
APA
Liang, J., Deng, X., Wu, F., & Tang, Y. (2013). Transcriptomic and proteomic analysis of human hepatic stellate cells treated with natural taurine. Molecular Medicine Reports, 7, 1442-1452. https://doi.org/10.3892/mmr.2013.1389
MLA
Liang, J., Deng, X., Wu, F., Tang, Y."Transcriptomic and proteomic analysis of human hepatic stellate cells treated with natural taurine". Molecular Medicine Reports 7.5 (2013): 1442-1452.
Chicago
Liang, J., Deng, X., Wu, F., Tang, Y."Transcriptomic and proteomic analysis of human hepatic stellate cells treated with natural taurine". Molecular Medicine Reports 7, no. 5 (2013): 1442-1452. https://doi.org/10.3892/mmr.2013.1389