Basic Research Open Access
Copyright ©The Author(s) 2003. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Sep 15, 2003; 9(9): 2050-2053
Published online Sep 15, 2003. doi: 10.3748/wjg.v9.i9.2050
Kangxian ruangan keli inhibits hepatic stellate cell proliferation mediated by PDGF
Ling Yang, Department of Traditional Chinese Medicine, Union Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
Chi-Zhi Zhang, Qing-Jing Zhu, Institute of Liver Diseases, Affiliated Hospital of Hubei College of Traditional Chinese Medicine, Wuhan 430061, Hubei Province, China
Author contributions: All authors contributed equally to the work.
Supported by the Natural Science Foundation of Hubei Province, No. 2000J042, and the Science Research Foundation of the Education Office of Hubei Province, No. 2000A06010
Correspondence to: Dr. Ling Yang, Department of Traditional Chinese Medicine, Union Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China. hepayang@163.com
Telephone: +86-27-85726395
Received: October 4, 2002
Revised: March 2, 2003
Accepted: March 12, 2003
Published online: September 15, 2003

Abstract

AIM: To investigate the effect of Kangxian ruangan keli (KXR) on hepatic stellate cell (HSC) proliferation mediated by platelet-derived growth factor (PDGF) and the underlying mechanism.

METHODS: In a serum-free culture system, HSCs were treated with a KXR preparation for 24 h, followed by stimulation with PDGF-BB for 24 h. Then the cells were incubated again in the medium containing KXR for 3 h stimulated with PDGF-BB for 5 minutes, and collected. The proliferation of HSC was examined using an MTT assay and flow cytometry. Tyrosine phosphorylation was detected with Western blotting and visualized by the enhenced chemiluminescent (ECL) method.

RESULTS: The OD values for the HSCs growing in the media without and with addition of PDGF were 0.17 ± 0.06 and 0.82 ± 0.05, respectively. The PDGF-induced increase was hindered remarkably by KXR preparation in a dose-dependent manner. The reaction values for the systems with 5 mg/mL, 2.5 mg/mL and 1.25 mg/mL of KXR were 0.28 ± 0.03, 0.37 ± 0.02 and 0.43 ± 0.04, respectively. Moreover, the percentages of S-phase cells in these KXR-containing culture systems were 10.95 ± 1.35, 32.76 ± 1.07 and 43.19 ± 1.09, respectively, all of which were significantly lower than that in the culture free of KXR (68.24 ± 2.72). In addition, the values for tyrosine-phosphorylated protein in HSCs treated with 5 mg/mL and 1.25 mg/mL of KXR were 0.1349 ± 0.0072 and 0.1658 ± 0.0025, respectively, which were smaller than that in the cells treated only with PDGF-BB (0.1813 ± 0.0117).

CONCLUSION: Within the dose range used in the present study, KXR preparation shows an inhibitory effect on HSC proliferation induced by PDGF. The mechanism of this process may involve interference with tyrosine phosphorylation mediated by PDGF.




INTRODUCTION

Hepatic fibrosis, a frequent pathologic change in a number of chronic liver diseases and an essential process during development of cirrhosis, is characterized by excessive proliferation of hepatic stellate cells (HSCs) and subsequent deposition of extracellular matrix (ECM)[1-6]. For this reason, HSC is regarded as one of the key cell types involved in progression of liver fibrosis, and is considered as a therapeutic target for treatment of hepatic fibrosis. Among several growth factors shown to be involved in the process, platelet-derived growth factor (PDGF) is the most potent mitogen for HSCs[7,8]. Hence, PDGF and its downstream target molecules or cell types may be potential targets for interventional therapies of hepatic fibrosis[9].

Kangxian ruangan keli (KXR) is an authorized granular herbal preparation, which has been used in the clinical fields for almost 30 years and is believed to be able to promote blood circulation, phlegm elimination and to soften and resolve hard mass based on traditional Chinese medicine. Our clinical and experimental data have demonstrated its effect on chronic liver diseases[10]. It may also be preventive to the fibrogensis induced by CCl4 and inhibitory to HSC proliferation[11-14]. However, its mechanism is unknown. In this study, the effect of KXR on PDGF-mediated HSC proliferation was assessed in vitro using a serum-free culture system.

MATERIALS AND METHODS
Reagents

HSC-T6, a cell line from activated HSCs, was kindly provided by Prof. Liemin Xu in Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine. DMEM medium and fetal calf serum were purchased from Gibco (Life Technologies, Inc., Gaithersburg, MD, USA). PDGF-BB, propidium iodide and Fura-2/AM were purchased from Sigma (St. Louis, MO, USA). The antibody to phosphotyrosine was purchased from Santa Cruz Biotechnology Inc. (Santa Cruz, CA, USA). The SuperSignal West Pico chemiluminescent substrate was purchased from Pierce Chemicals (Rockford, IL, USA).

Cell culture

HSCs were grown in DMEM medium containing 100 mL/L fetal calf serum, 1 × 10-5 U/L penicillin, 100 mg/L streptomycin, 1% L-glutamine and 0.1 mmol/L HEPES, and cultured at 37 °C in an incubator with 5% CO2. KXR consists of several berbs including Radix Salviae Miltiorrhizae, Rhizoma Zedoariae, Sargassum, Carapax Trionycis[15]. The granular preparation was made in the Affiliated Hospital of Hubei Collage of Traditional Chinese Medicine, containing 2 g of crude herbs in each gram KXR. The preparation was dissolved in DMEM medium, and sterilized by filtration through a 0.45 μm filter. Following preincubation in the serum-free medium for 24 h, the cells were grown and incubated for 48 h, separately in the following media: 1) Only serum-free DMEM medium as a control, 2) The serum-free medium containing 10 ng/mL PDGF-BB, 3) before the addition of PDGF-BB (10 ng/mL), different amounts of KXR (1.25 mg/mL, 2.5 mg/mL and 5 mg/mL, separately) were extracted in the serum-free medium for 3 h.

Evaluation of cell growth

Cells were seeded in 96-well plates at a density of 1 × 108 cells/L. After treated as above, the cells were incubated with DMEM containing 1 mg/mL of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) for 4 h at 37 °C. The resulted precipitate was resolved by addition of dimethyl sulfoxide and incubation for 2 min. Cell growth was assessed using an enzyme-linked immunosorbent assay reader (Bio-Tek, Houston, USA) with the test wavelength at 570 nm and expressed as optical density (OD) values. The inhibition of HSC proliferation was expressed as inhibitory rate [(OD value of PDGF group - OD value of KXR group)/OD value of PDGF group] × 100%.

The cell proliferation was also described by the growth curves. HSCs were plated in 24-well dishes at a density of 1 × 105 cells/well in a complete culture medium. After 24 h (day 0), cells were washed twice with the serum-free medium and then divided and treated as described above. Cell counting was performed on triplicate wells on days 0, 2 and 4 following preparation of cell suspensions by digestion with trypsin. Fresh serum-free medium containing PDGF-BB (10 ng/mL) with or without KXR was added to the remaining wells on day 2.

The HSC proliferation kinetics was described by flow cytometry. The cells were plated in 10-cm dishes at a density of 1 × 108 cells/L in the complete culture medium. After treated as described above, cells were harvested by digestion with trypsin and washed twice with PBS. After overnight fixation with cold ethanol at 4 °C, cells were washed twice with PBS and suspended in the buffer containing 30 μg/mL propidium iodide, 0.1% Tritonx-100, 37 mg/mL EDTA and 0.1% ribonuclease A. Samples were incubated in a dark chamber at room temperature for 30 minutes and stored at 4 °C before use. Cell fluorescence was measured by FACSCalibur flow cytometer (Becton Dickson, San Diego, CA, USA) and assessed by the Cellquest software.

Tyrosine phosphorylation assay

Subconfluent HSCs were cultured in a serum-free medium for 24 h, then in the medium with or without KXR for another 24 h. Following the culture with the presence of 10 ng/mL PDGF-BB for 24 h, cells were treated with KXR for 3 h, and incubated again in the medium containing 10 ng/mL PDGF-BB for 5 min. After washed with ice-cold PBS, the cell monolayers were harvested and suspended in a lysis buffer [1% NP-40, 0.5% sodium deoxycholate, 0.1% sodium dodecyl sulfate (SDS), 150 mmol/L NaCl, 1 mmol/L ethyleneglycol-bis-(β-aminoethylether)-N, N, N’, N’-tetraacetic acid, 1 mmol/L phenylmethylsulfonyl fluoride, 500 μg/mL leupepin, 1 mmol/L sodium orthovanadate, 50 mmol/L sodium fluorid, 10 mmol/L Tris-HCl, pH7.2[16]] for 30 min on ice. The lysates were clarified by centrifugation at 16000 g for 10 min at 4 °C, and protein concentrations were determined using Bradford’s method[16].

Proteins were electrophoresed on a 0.1% SDS-7% polyacrylamide gel using a Mini apparatus (Bio-Rad Laboratories, Hercules, CA, USA). The resolved proteins were transferred to a nitrocellulose filter by a transfer apparatus (Bio-Rad Laboratories, Hercules, CA, USA). After the blocking in a blocking buffer [5% BSA, 1% hen egg albumin, 10 mmol/L Tris.Cl, pH7.4, 0.15 mol/L NaCl] for 60 min, the filter was incubated with a mouse monoclonal antibody to phosphotyrosine (1:1000) for 120 min. Following the washing for 15 minutes respectively in a TN buffer (containing 10 mmol/L Tris pH7.4, 0.15 mol/L NaCl) and a TN buffer containing 0.05% NP-40, the incubation was done with a peroxidase-conjugated goat antibody (1:2000) against mouse IgG at room temperature for 60 min. The antigen-antibody complexes were visualized by an enhanced chemiluminescent (ECL) reaction[16]. The reaction intensities were assessed with an image analyzer (HPIAS-1000, Tongji Medical college Huazhong University of Science & Technology, Wuhan, China).

Statistical analysis

Results were expressed as mean ± standard deviation. Differences between groups were described by the Student t test. P < 0.05 was considered significant.

RESULTS
Effect of KXR on PDGF-mediated HSC Proliferation

An inhibitory effect was observed by the MTT assay (Table 1) and cell counting (Figure 1) for KXR to PDGF-mediated HSC proliferation, which was in a dose-dependent manner. HSC growth response to PDGF stimulation with or without the presence of KXR was assessed by cell counting (Figure 1). PDGF-BB, with its concentration at 10 ng/mL, significantly increased HSC growth after 2 to 4 days of incubation, in comparison to the medium free of PDGF (P < 0.01). This response was markedly inhibited by pretreatment with KXR (P < 0.01), which was not associated with any detectable cytotoxic effects in the dose range used.

Table 1 Effects of KXR on PDGF-mediated HSC proliferation (mean ± s).
GroupsKXR concentration (mg/mL)OD valuesInhibitory rate (%)
Control0.17 ± 0.06b-
PDGF0.82 ± 0.050.00
KXR treatment before50.28 ± 0.03b65.9
PDGF stimulation2.50.37 ± 0.02b54.9
1.250.43 ± 0.04b47.6
Figure 1
Figure 1 Effect of Kangxian Ruangan Granule on PDGF-induced cell proliferation in HSC-T6 cell line.
KXR affects cell cycle of HSC

Flow cytometry showed a reduction in G0/G1-phase cell fraction and an increase in the S-phase cell fraction for the HSCs exposed to PDGF. Pretreatment with KXR inhibited the PDGF-mediated response for the cells in S phase, the result was in agreement with the data obtained by MTT assay. However, the G0/G1-phase cell fraction was similar to that without KXR pretreatment (Table 2). It indicated that KXR treatment was inhibitory to the PDGF-induced progression of the cell cycle beyond the G1 phase. The effect seemed to be dose-dependent (P < 0.01).

Table 2 Effect of KXR on cell cycle progression of HSCs mediated by PDGF (mean ± s).
GroupsnHSC fractions at (%)
G0-G1 phaseS phaseG2-M phase
Control694.67 ± 2.174.13 ± 0.921.20 ± 0.47
PDGF 10 ng/mL620.18 ± 1.1268.24 ± 2.7211.18 ± 1.93
KXR 5 mg/mL→PDGF683.64 ± 3.68b10.95 ± 1.35b5.41 ± 0.98b
KXR 2.5 mg/mL→PDGF662.58 ± 4.52b32.76 ± 1.07b4.66 ± 0.81b
KXR 1.25 mg/mL→PDGF648.18 ± 3.37b43.19 ± 1.09b8.63 ± 0.71a
Effect of KXR on PDGF-induced tyrosine phosphorylation

As shown in Figure 2 and Table 3, treatment of KXR resulted in a reduction in the content of tyrosine-phosphorylated proteins, the result was in accordance with its effects on cell proliferation and kinetics.

Figure 2
Figure 2 Effect of KXR on the expression of tyrosine-phosphorylated proteins induced by PDGF (Western blot). Lanes 1 to 4 were loaded with HSC lysates from the control, PDGF groups, and the PDGF groups with pretreatment of KXR at 1.25 mg/mL and 5 mg/mL, respectively.
Table 3 Effect of KXR on the contents of tyrosine-phosphorylated proteins in HSCs stimulated by PDGF (mean ± s).
GroupsnIntensities of tyrosine-phosphorylated proteins
Control40.1199 ± 0.0037
PDGF 10 mg/mL40.1813 ± 0.0117
KXR 1.25 mg/mL+PDGF40.1658 ± 0.0025a
KXR 5 mg/mL+PDGF40.1349 ± 0.0072b
DISCUSSION

KXR is a granular herb preparation mainly composed of Salviae Miltiorrhizae, Rhizoma Zedoariae, Sargassum and Carapax Trionycis. It has been reported that Salviae Miltiorrhizae exhibits a series of important effects including anti-inflammation, anti-oxidation and retardation of HSC proliferation[17,18]. Its extract has been shown to induce HSC apoptosis[19,20]. The oil from Rhizoma Zedoariae was reported to be inhibitory to the proliferation of fibroblast cells and hepatocellular carcinoma cells[21,22]. It was pointed out that Sargassum could be helpful for the elimination of reactive oxygen species which were believed to be important factors in the progression of fibrosis[23]. Our previous clinical and experimental observations[10-14] have approved these effects using decoction preparation of KXR in patients with chronic hepatitis or cirrhosis and in rats with CCL4-induced liver fibrosis.

HSCs are a key cell type during liver fibrogenesis. Its activation is characterized by the myofibroblast-like phenotypes including an elevated proliferation rate, expression of α-smooth muscle actin, synthesis and excretion of some extracellular matrix components[4,24]. Recently, the phenotypic transformation has been linked to some cytokines, including PDGF, and their intracellular signal transduction pathways[6,25-30].

Among many polypeptide growth factors potentially involved in chronic hepatitis, PDGF, a dimmer of two chains referred to as A-chain and B-chain, has been shown to be the most potent mitogen for cultured HSCs isolated from rat, mouse or human liver. Of the three possible dimeric forms, including AA, AB and BB, PDGF-BB has been shown to be most effective[1,6-8,30]. Tyrosine protein kinase plays an important role in the PDGF-mediated activation and proliferation of HSCs[32,33]. PDGF binds to the extracellular domain of its receptor, PDGF-Rβ, results in the receptor molecule autophosphorylation, which allows the docking of numerous signaling cascades, such as PI3-K, Raf-MEK-MAPK, Na+/H+ and Ca2+, leading to cell division and chemotaxis[6,34-38]. In the current study, we showed that KXR blocked PDGF-stimulated mitogenesis and DNA synthesis, and inhibited PDGF-induced autophosphorylation of tyrosine. These results suggest KXR may have the effects as those of tyrosine phosphorylation inhibitors. Clearly, more data are needed to elucidate the effect of KXR on tyrosine phosphorylation and its underlying mechanism.

Footnotes

Edited by Su Q

References
1.  Friedman SL. Cytokines and fibrogenesis. Semin Liver Dis. 1999;19:129-140.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 265]  [Cited by in F6Publishing: 259]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
2.  Jiang HQ, Zhang XL. Progress in the study of pathogenesis in hepatic fibrosis. Shijie Huangren Xiaohua Zazhi. 2000;8:687-689.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Wang JY, Zhang QS, Guo JS, Hu MY. Effects of glycyrrhetinic acid on collagen metabolism of hepatic stellate cells at different stages of liver fibrosis in rats. World J Gastroenterol. 2001;7:115-119.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Reeves HL, Friedman SL. Activation of hepatic stellate cells--a key issue in liver fibrosis. Front Biosci. 2002;7:d808-d826.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 319]  [Cited by in F6Publishing: 353]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
5.  Pinzani M, Marra F, Carloni V. Signal transduction in hepatic stellate cells. Liver. 1998;18:2-13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 172]  [Cited by in F6Publishing: 182]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
6.  Marra F, Gentilini A, Pinzani M, Choudhury GG, Parola M, Herbst H, Dianzani MU, Laffi G, Abboud HE, Gentilini P. Phosphatidylinositol 3-kinase is required for platelet-derived growth factor's actions on hepatic stellate cells. Gastroenterology. 1997;112:1297-1306.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 161]  [Cited by in F6Publishing: 168]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
7.  Kinnman N, Goria O, Wendum D, Gendron MC, Rey C, Poupon R, Housset C. Hepatic stellate cell proliferation is an early platelet-derived growth factor-mediated cellular event in rat cholestatic liver injury. Lab Invest. 2001;81:1709-1716.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 92]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
8.  Iwamoto H, Nakamuta M, Tada S, Sugimoto R, Enjoji M, Nawata H. Platelet-derived growth factor receptor tyrosine kinase inhibitor AG1295 attenuates rat hepatic stellate cell growth. J Lab Clin Med. 2000;135:406-412.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 32]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
9.  Bataller R, Brenner DA. Hepatic stellate cells as a target for the treatment of liver fibrosis. Semin Liver Dis. 2001;21:437-451.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 373]  [Cited by in F6Publishing: 385]  [Article Influence: 16.7]  [Reference Citation Analysis (0)]
10.  Zhang CZ, Yan HM, Wang L. Effect of Kangxian Ruangan Chongji on 31 cases of liver cirrhosis. Zhongxiyi Jiehe Ganbing Zazhi. 1999;2:19-20.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Xiong YQ, Yan HM, Zhang CZ. Experimental study of the Kangxian Ruangan granute on anti-hepatic fibrosis in rats. Zhongguo Shiyan Fangjixue Zazhi. 2000;6:28-30.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Yang L, Zhu QJ, Zhang CZ. Serapharmlocogical effect of Kangxian Ruangan Chongji on expression of type I procollagen and transforming growth factor β1 mRNA in hepatic stellate cells. Zhongguo Zhongyi Jichu Yixue Zazhi. 2001;8:38-40.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Yang L, Cheng HQ, Zhu QJ, Zhang CZ. Effects of Kangxian Ruangan Granules on platelet-derived growth factor (PDGF) in-duced cell cycle and intercellular calium concentration of hepatic stellate cells. Zhongxiyi Jiehe Ganbing Zazhi. 2002;1:20-22.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Zhou Z, Zhang C, Chen J, Zhou P. [Serapharmacological effect of kangxian ruangan granula on the proliferation of hepatic stellate cells]. Zhongyaocai. 2001;24:809-811.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Feng H, Zhang H, Tang D. Study on preparation and quality of Kangxian Ruangan Granule. Zhongxiyi Jiehe Ganbing Zazhi. 2000;4:31-32.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Ausubel F. Short protocols in molecular biology. 1st ed. Beijing: Science Publishing House. 1999;687-693.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Liu CH, Liu P, Hu YY, Xu LM, Tan YZ, Wang ZN, Liu C. Effects of salvianolic acid-A on rat hepatic stellate cell proliferation and collagen production in culture. Acta Pharmacol Sin. 2000;21:721-726.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Liu P, Liu CH, Wang HN, Hu YY, Liu C. Effect of salvianolic acid B on collagen production and mitogen-activated protein kinase activity in rat hepatic stellate cells. Acta Pharmacol Sin. 2002;23:733-738.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Zhang XL, Liu L, Jiang HQ. Salvia miltiorrhiza monomer IH764-3 induces hepatic stellate cell apoptosis via caspase-3 activation. World J Gastroenterol. 2002;8:515-519.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Yao XX, Tang YW, Yao DM, Xiu HM. Effects of Yigan Decoction on proliferation and apoptosis of hepatic stellate cells. World J Gastroenterol. 2002;8:511-514.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Shi LC, Wu WY, Zhang WB, Qu YQ, Tan M, Xiao CM. Effect of curcuma aramatica oil on proliferating cell nuclear antigen of hepatoma in mice. Shijie Huangren Xiaohua Zazhi. 1999;7:156-157.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Xin JJ, Li GY, Cui KY. Effects of algae-selenium polysaccharide on immune function of mice. Zhongguo Haiyang Yaowu. 1999;3:36-38.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Zheng G, Hao J, Jia GR, Zhong HM, Song YM. Effects of algin on preventing liver fibrosis in rat. Zhongguo Haiyang Yaowu. 1997;1:30-32.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Pinzani M, Marra F. Cytokine receptors and signaling in hepatic stellate cells. Semin Liver Dis. 2001;21:397-416.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 330]  [Cited by in F6Publishing: 326]  [Article Influence: 14.2]  [Reference Citation Analysis (0)]
25.  Iredale JP. Hepatic stellate cell behavior during resolution of liver injury. Semin Liver Dis. 2001;21:427-436.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 250]  [Cited by in F6Publishing: 246]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
26.  Shen H, Huang GJ, Gong YW. Effect of transforming growth factor beta and bone morphogenetic proteins on rat hepatic stellate cell proliferation and trans-differentiation. World J Gastroenterol. 2003;9:784-787.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Huang GC, Zhang JS. Signal transduction in activated hepatic 38 stellate cells. Shijie Huaren Xiaohua Zazhi. 2001;9:1056-1060.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Britton RS, Bacon BR. Intracellular signaling pathways in stellate cell activation. Alcohol Clin Exp Res. 1999;23:922-925.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 28]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
29.  Maeda N, Kawada N, Seki S, Arakawa T, Ikeda K, Iwao H, Okuyama H, Hirabayashi J, Kasai K, Yoshizato K. Stimulation of proliferation of rat hepatic stellate cells by galectin-1 and galectin-3 through different intracellular signaling pathways. J Biol Chem. 2003;278:18938-18944.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 111]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
30.  Tahashi Y, Matsuzaki K, Date M, Yoshida K, Furukawa F, Sugano Y, Matsushita M, Himeno Y, Inagaki Y, Inoue K. Differential regulation of TGF-beta signal in hepatic stellate cells between acute and chronic rat liver injury. Hepatology. 2002;35:49-61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 158]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
31.  Pinzani M. PDGF and signal transduction in hepatic stellate cells. Front Biosci. 2002;7:d1720-d1726.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Liu XJ, Yang L, Mao YQ, Wang Q, Huang MH, Wang YP, Wu HB. Effects of the tyrosine protein kinase inhibitor genistein on the proliferation, activation of cultured rat hepatic stellate cells. World J Gastroenterol. 2002;8:739-745.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Carloni V, Pinzani M, Giusti S, Romanelli RG, Parola M, Bellomo G, Failli P, Hamilton AD, Sebti SM, Laffi G. Tyrosine phosphorylation of focal adhesion kinase by PDGF is dependent on ras in human hepatic stellate cells. Hepatology. 2000;31:131-140.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 56]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
34.  Marra F, Arrighi MC, Fazi M, Caligiuri A, Pinzani M, Romanelli RG, Efsen E, Laffi G, Gentilini P. Extracellular signal-regulated kinase activation differentially regulates platelet-derived growth factor's actions in hepatic stellate cells, and is induced by in vivo liver injury in the rat. Hepatology. 1999;30:951-958.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 128]  [Cited by in F6Publishing: 132]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
35.  Carloni V, Defranco RM, Caligiuri A, Gentilini A, Sciammetta SC, Baldi E, Lottini B, Gentilini P, Pinzani M. Cell adhesion regulates platelet-derived growth factor-induced MAP kinase and PI-3 kinase activation in stellate cells. Hepatology. 2002;36:582-591.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 36]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
36.  Reeves HL, Thompson MG, Dack CL, Burt AD, Day CP. The role of phosphatidic acid in platelet-derived growth factor-induced proliferation of rat hepatic stellate cells. Hepatology. 2000;31:95-100.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 21]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
37.  Di Sario A, Bendia E, Svegliati Baroni G, Ridolfi F, Bolognini L, Feliciangeli G, Jezequel AM, Orlandi F, Benedetti A. Intracellular pathways mediating Na+/H+ exchange activation by platelet-derived growth factor in rat hepatic stellate cells. Gastroenterology. 1999;116:1155-1166.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 39]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
38.  Di Sario A, Bendia E, Taffetani S, Marzioni M, Candelaresi C, Pigini P, Schindler U, Kleemann HW, Trozzi L, Macarri G. Selective Na+/H+ exchange inhibition by cariporide reduces liver fibrosis in the rat. Hepatology. 2003;37:256-266.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 40]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]