Electroporative interleukin-10 gene transfer ameliorates carbon tetrachloride-induced murine liver fibrosis by MMP and TIMP modulation
Full-length article

Electroporative interleukin-10 gene transfer ameliorates carbon tetrachloride-induced murine liver fibrosis by MMP and TIMP modulation1

Wen-ying Chou2, Cheng-nan Lu3, Tsung-hsing Lee2, Chia-ling Wu2,5, Kung-sheng Hung4, Allan M Concejero4, Bruno Jawan2, Cheng-haung Wang2,6

Departments of 2Anesthesiology, 3Chinese Medicine and 4Surgery, Kaohsiung Chang Gung Memorial Hospital, Taipei, China; 5Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, China

1Project supported by research grant CMRPG- 840451 from the Chang Gung Memorial Hospital.

6Correspondence to Dr Cheng-haung WANG.
Phn 86-7-731-7123, ext 8746. Fax 86-7-711-2776.
E-mail wang1211@adm.cgmh.org.tw


Aim: Liver fibrosis represents a process of healing and scarring in response to chronic liver injury. Effective therapies for liver fibrosis are lacking. Interleukin-10 (IL-10) is a cytokine that downregulates pro-inflammatory responses and has a modulatory effect on hepatic fibrogenesis. The aim of this study was to investigate whether electroporative IL-10 gene therapy has a hepatic fibrolytic effect on mice.

Methods: Hepatic fibrosis was induced by administering carbon tetrachloride (CCl4) for 10 weeks in mice. The human IL-10 expression plasmid was delivered via electroporation after hepatic fibrosis was established. Histopathology, reverse transcription polymerase chain reaction (RT-PCR), immunoblotting, and gelatin zymography were used to investigate the possible mechanisms of action of IL-10.

Results: Human IL-10 gene therapy reversed CCl4-induced liver fibrosis in mice. RT-PCR revealed that IL-10 gene therapy attenuated liver TGF-β1, collagen α1, fibronectin, and cell adhesion molecule mRNA upregulation. Following gene transfer, both the activation of α-smooth muscle actin and cyclooxygenase-2 were significantly attenuated. Furthermore, IL-10 significantly inhibited matrix metalloproteinase-2 (MMP-2) and tissue inhibitors of matrix metalloproteinase (TIMP) activation after CCl4 intoxication.

Conclusions: We demonstrated that IL-10 gene therapy attenuated CCl4-induced liver fibrosis in mice. IL-10 prevented upregulated fibrogenic and pro-inflammatory gene responses. Its collagenolytic effect may be attributed to MMP and TIMP modulation. IL-10 gene therapy may be an effective therapeutic modality against liver fibrosis with potential clinical use.

Keywords: interleukin-10; gene therapy; liver cirrhosis; gelatinase A; tissue inhibitor of metalloproteinases; cyclooxygenase 2


Submitted Oct 09, 2005. Accepted for publication Dec 23, 2005.

doi: 10.1111/j.1745-7254.2006.00304.x


Introduction

Oxidative stress, an important factor that induces liver fibrosis, represents a key feature of hepatitis induced by various conditions, including anoxic/reoxygenation injury, autoimmune hepatitis, viral hepatitis and alcoholic hepatitis[1]. Less severe oxidative stress may sustain fibrosis progression by causing activation and morphological changes in hepatic stellate cells (HSC), including promoting proliferative activity, synthesis and degradation/remodeling of the extracellular matrix (ECM), chemotaxis, contractility, proinflammatory activity and retinoid loss[1,2].

Carbon tetrachloride (CCl4) is a xenobiotic used extensively to induce oxidative stress. It is assumed to initiate free radical-mediated lipid peroxidation, leading to the accumulation of lipid-derived oxidation products that cause liver injury and excess collagen deposition in the liver, resulting in liver fibrosis[3,4]. During hepatic fibrogenesis, there is an imbalance between excess synthesis of ECM and/or its removal, with consequent fibrosis and scarring[5,6]. The pathophysiology of ECM formation during liver fibrosis is multifaceted and complex[7,8]. It involves a change in the expression of ECM proteases (matrix metalloproteinases; MMP) or their inhibitors (tissue inhibitors of metalloproteinases; TIMP) and an increase in the synthesis of collagen and fibronectin driven by signaling pathways mediated by pro-inflammatory cytokines such as transforming growth factor-β1 (TGF-β1) and tumor necrosis factor-α (TNF-α)[912].

Interleukin (IL)-10 is a cytokine that downregulates pro-inflammatory responses[13]. Human IL-10 is a 160 amino acid protein (molecular weight=18.5 kDa), and murine IL-10 is a 157 amino acid protein with 80% homology to the human form[14]. Recombinant human IL-10 has been produced and tested in clinical trials. Studies suggest that IL-10 may be effective against chronic hepatitis C and other liver diseases[15]. Further, IL-10 gene therapy has been studied extensively in animal models for autoimmune diabetes, thyroiditis, and colitis[1618]. Because the elimination half-life of recombinant IL-10 is relatively short (=2 h)[19], it may be possible to utilize its therapeutic properties to develop a gene-based treatment regimen. In the present study, we investigate whether IL-10 gene therapy is effective against CCl4-induced liver fibrosis in mice.


Materials and methods

Subjects Male 6- to 8-week-old ICR mice were purchased from the National Science Council, Taiwan, China, and were allowed to acclimatize for 5 d before experimentation. The mice were housed in Kaohsiung Chang Gung Memorial Hospital Animal Facility under standard temperatures, and with a standard light and dark cycle. All procedures performed on the mice were approved by the Kaohsiung Chang Gung Memorial Hospital Animal Care and Use Committee.

IL-10 expression plasmid preparation A human IL-10 expression plasmid (pCYIL-10 vector) was used in the present study[20]. In brief, full-length human IL-10 cDNAs were subcloned into a pCY4B expression vector driven by a chicken β-actin promoter with a cytomegalovirus immediate early enhancer. pCMV-Lacz was used as the vehicle control. These plasmids were purified using the EndoFree Plasmid Giga Kit (Qiagen, Valencia, CA, USA).

Liver fibrosis induction and gene therapy Based on the method used in a previous study, but with some modifica-tions, the mice were administered CCl4 (1 mL/kg body weight) dissolved in olive oil (1:1) twice a week for 10 weeks[21]. Sixteen mice were killed at the end of 6 weeks to confirm that liver fibrosis was established (group I). To evaluate the anti-fibrotic effect of IL-10, gene therapy administration was started at the end of 6 and 8 weeks of CCl4 treatment. Briefly, 30 µL bovine hyaluronidase (0.4 IU/µL) (Sigma-Aldrich, St Louis, MO, USA) was injected into the anterior tibialis (AT) muscle of the mice 2 h before electroporation. pCYIL-10 was injected into the bilateral AT muscles using a 27G needle (30 µL into each leg; 4 µg/µL; group II, n=16). Electroporation was carried out using electrical pulses (8 pulses of 20 ms, 175 V/cm, and 1 s intervals) with Tweezertrode electrode disks and an electrical pulse generator (T830; BTX, San Diego, CA, USA)[22].

Sixteen mice received gene electro-transfer therapy using the same procedure as described above using pCMV-LacZ (group III) as a vehicle control at the end of 6 and 8 weeks. All surviving mice (group II, n=12; group III, n=7) were killed at the end of the 10-week CCl4 treatment. Five mice were killed before CCl4 intoxication as normal controls (group N).

Histopathology and immunohistochemistry For histopathology studies, mice were killed at 0, 6, and 10 weeks after CCl4 administration. The liver was removed and fixed in 10% formalin solution. Five-micrometer sections were stained with 0.1% Sirius red in picric acid (Sigma-Aldrich). Matrix density was quantified using a computerized image analysis system as previously described[23]. For immunohistochemical studies, the sections were washed in phosphate-buffered saline (PBS), and incubated in 3% normal goat serum with 0.3% Triton X-100 in PBS for 1 h. The sections were incubated free-floating at 4 °C with IL-10 (specific for human origin; Santa Cruz Biotechnology, Santa Cruz, CA, USA), cyclooxygenase-2 (COX-2), MMP-2, and TIMP-1 (Abcam, Cambridge, MA, USA) antibodies. Immunoreactivity was visualized using the Vectastain Elite ABC Peroxidase method (Vector Laboratories, Burlingame, CA, USA) with diamino-benzidine (DAB) as the chromagen.

Soluble collagen measurement For soluble collagen analysis, the Sircol collagen assay (Biocolor, Belfast, UK) was performed following the manufacturer’s instructions as described in a previous study[24]. Briefly, 50 mg of liver was homogenized. Total acid pepsin-soluble collagens were extracted overnight using 5 mg/mL pepsin in 500 µL of 0.5 mol/L acetic acid. One milliliter of Sircol dye reagent was added to every 100 µL of each sample, in duplicate, and the mixture was incubated at 25 °C for 30 min. After centrifugation, the pellet was suspended in 1 mL of alkali reagent. The absorbance was read at 540 nm.

Immunoblotting The liver specimens were homogenized in a lysis buffer with complete protease inhibitor cocktail tablets (Roche, Mannheim, Germany). For analysis of α-smooth muscle actin (α-SMA) expression after CCl4 administration, 20 μg of protein extracts were electrophoresed on a 10% acrylamide sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) gel and immunoblotted onto PVDF membranes. The membranes were blocked for 1 h at room temperature and incubated overnight with a 1:1000 dilution of α-SMA, and α-tubulin antibodies (Abcam). Antibody binding was detected using horseradish peroxidase (HRP)-linked immunoglobulin G (IgG). Bands were visualized using an ECL detection system (Amersham-Pharmacia Biotech, Little Chalfont, UK). Band intensities were quantified using an image analyzer (Densitograph AE-6900M; Atto, Tokyo, Japan).

Reverse transcription-polymerase chain reaction Livers were harvested at 0, 6, and 10 weeks after CCl4 admini-stration. The expression levels of TGF-β1, collagen α1, fibronectin, TNF-α, intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), TIMP-1, and TIMP-2 mRNA were analyzed using reverse transcription-polymerase chain reaction (RT-PCR) techniques. The total RNA was extracted and then reverse-transcribed into cDNA. PCR was performed at a final concentration of 1× PCR buffer, 1.0 µmol/L of each of the 3' and 5' primers, and 10 U of Advan-Taq Plus DNA polymerase (Clontech, Palo Alto, CA, USA) in a total volume of 50 µL. The mixture was amplified for 32 cycles in a thermal cycler (Stratagene, La Jolla, CA, USA). The β-actin was amplified to verify equal loading. The primer sequence and expected product size were as previously described[25]. The amplification products were separated by agarose gel electrophoresis and visualized using ethidium bromide staining. The gel was scanned at a NucleoVision imaging workstation (NucleoTech, San Mateo, CA, USA), and quantified using GelExpert release 3.5.

Gelatin zymography Gelatin zymography was carried out to explore MMP activity. Briefly, liver tissues were homogenized in a protein extraction buffer. The supernatant of a centrifuged liver sample (20 µg of protein extract per line) was mixed 1:3 with a sample buffer and separated by sodium dodecylsulfate (SDS)-polyacrylamide gel electrophoresis in 8% polyacrylamide gel copolymerized with 1 mg/mL gelatin (Sigma-Aldrich) as described elsewhere[26]. Gels were incubated at 37 °C overnight in an MMP activation buffer. After Coomassie blue staining, the extent of gel digestion localized to bands of active-MMP-2 (64-kDa) were quantified by densitometry.

Statistical analysis All data (from at least 3 separate experiments) are presented as mean±SEM. Statistical analysis was performed using one-way ANOVA followed by the t-test. P<0.05 was considered significant.


Results

Long-term IL-10 expression following electroporative gene transfer There was only scanty staining of cells for human IL-10 in the non-gene transfer groups (groups N, I, and III; Figure 1). Strong positive staining of cells for human IL-10 was seen in the livers of the gene transfer group II and 4 weeks after electroporation (group II; Figure 1C).

Figure 1 Representative photomicrographs of IL-10-stained mouse liver sections. There was scanty staining for human IL-10 in the non-gene therapy groups (groups N, I, and III). Strong positive staining for human IL-10 was detected in the livers of the gene transfer group (group II). (A) Normal control; (B) Group I; (C) Group II; (D) group III. Arrows indicate IL-10-positive cells. Magnification, ×400 (inset, ×200).

IL-10 gene therapy reversed CCl4-induced liver fibrosis There was no significant difference in food and water intake throughout the study period between groups. After 6 weeks of CCl4 administration, liver fibrosis was seen histo-pathologically. Sirius red staining of liver sections revealed extensive fibrosis, portal-to-portal fibrous bridging, and nodular transformation in groups I and III (Figure 2). Human IL-10 gene therapy (group II) significantly ameliorated hepatic fibrogenesis and reduced matrix density (Figure 2C). These findings were further confirmed by measurements of liver collagen content (Table 1).

Figure 2 Representative photomicrographs of Sirius red-stained mouse liver sections. Extensive fibrosis, portal-to-portal fibrous bridging, and nodular transformation were seen in groups I and III after chronic CCl4 administration. IL-10 gene therapy (group II) significantly reduced matrix density and abrogated hepatic fibrogenesis. (A) Normal control; (B) Group I; (C) Group II; (D) Group III. Magnification, ×50.
Table 1
Table 1 Effects of IL-10 gene therapy on regression of hepatic fibrosis. IL-10 gene therapy significantly reduced the degree of liver fibrosis caused by chronic CCl4 administration in mice as measured using Sirius red matrix density and collagen content. Mean±SEM. bP<0.05,cP<0.01 vs group I. fP<0.01 vs group III.
Full table

IL-10 gene therapy attenuated COX-2 increment after CCl4 COX-2 was not detected immunohistochemically in the normal group. COX-2 expression was upregulated after CCl4 administration (groups I and III; Figure 3). IL-10 gene therapy significantly diminished this COX-2 expression (Figure 3C).

Figure 3 Representative photomicrographs of COX-2-stained mouse liver sections. COX-2 was not detected in the normal control group. COX-2 expression was increased in groups I and III after CCl4 administration. IL-10 gene therapy (group II) significantly attenuated this increase. (A) Normal control; (B) Group I; (C) Group II; (D) Group III. Arrows indicate COX-2 positive cells. Magnification ×400 (inset ×200).

IL-10 gene therapy suppressed hepatic stellate cell activation after CCl4 α-SMA [activated hepatic stellate cell (HSC) markers] are known to be activated after acute liver injury[27,28]. In the present study, the expression of α-SMA increased after chronic CCl4 administration as measured using immunoblotting (Figure 4). IL-10 gene therapy (group II) significantly reduced this upregulation, indicating HSC inactivation (P<0.01 vs group I; P<0.01 vs III). α-Tubulin was used as an internal control.

Figure 4 Representative photomicrographs of COX-2-stained mouse liver sections. COX-2 was not detected in the normal control group. COX-2 expression was increased in groups I and III after CCl4 administration. IL-10 gene therapy (group II) significantly attenuated this increase. (A) Normal control; (B) Group I; (C) Group II; (D) Group III. Arrows indicate COX-2 positive cells. Magnification ×400 (inset ×200).

IL-10 gene therapy attenuated fibrogenic, proinflamma-tory, and cell adhesion molecule gene responses after CCl4 treatment Expression of TGF-β1, collagen α1, fibronectin, TNF-α, ICAM-1, and VCAM-1 mRNA were all upregulated in the fibrotic liver as semi-quantified using RT-PCR (Figure 5). β-actin was amplified as an internal control. IL-10 gene therapy (group II) significantly attenuated these increase. In brief, IL-10 gene transfer suppressed the fibrogenic, pro-inflammatory, and cell adhesion molecule gene responses after CCl4 administration.

Figure 5 Representative photographs of semi-quantified RT-PCR in the CCl4-treated mouse liver. IL-10 gene therapy (group II) significantly attenuated TGF-β1, collagen α1, fibronectin, TNF-α, ICAM-1, VCAM-1, TIMP-1, and TIMP-2 mRNA activation after CCl4 administration when compared with groups I and III (bP<0.05, cP<0.01 vs group I. fP<0.01 vs group III). β-Actin was amplified to verify equal loading. The arbitrary units are defined as target gene/β-actin. Mean±SEM.

IL-10 gene therapy attenuated MMP-2 activation in the fibrotic liver The expression of MMP after CCl4 treatment was evaluated by using immunohistochemical and gelatin zymography methods. Immunohistochemical studies showed that when compared with normal livers, MMP-2 levels were significantly increased in the fibrotic livers (groups I and III; Figure 6). IL-10 gene therapy attenuated this upregulation (group II; Figure 6C). The collagenolytic activity of MMP protein in liver homogenates was examined by zymography (Figure 7). Gelatin zymography showed that the concentration of the 64 kDa active MMP-2 molecule increased in groups I and III after CCl4 administration, and IL-10 gene therapy (group II) abrogated this increase (P<0.01).

Figure 6 Representative photomicrographs of MMP-2-stained mouse liver sections. After CCl4 administration, MMP-2 staining was significantly accentuated in groups I and III. IL-10 gene therapy (group II) decreased this upregulation. (A) Normal control; (B) Group I; (C) Group II; (D) Group III. Arrows indicate MMP-2-positive cells. Magnification, ×400 (inset, ×200).
Figure 7 Representative photograph of gelatin zymography in CCl4-treated mouse liver. When compared with the normal control group, levels of the active 64 kDa MMP-2 molecule were increased in groups I and IV after CCl4 treatment. IL-10 gene therapy attenuated MMP-2 activation significantly (cP<0.01 vs group I. fP<0.01 vs group III).

IL-10 gene therapy attenuated TIMP activation after CCl4 treatment Expression of TIMP in the fibrotic livers was also evaluated by RT-PCR and immunohistochemical methods. RT-PCR showed that TIMP-1 and TIMP-2 mRNA were significantly upregulated in the fibrotic liver. IL-10 gene therapy (group II) significantly attenuated these increase (P<0.01, Figure 5). Immunohistochemical studies revealed that the level of TIMP-1 was increased after chronic CCl4 administration (Figure 8). IL-10 gene therapy (group II) significantly attenuated this activation (Figure 8C).

Figure 8 Representative photomicrographs of TIMP-1-stained mouse liver sections. After CCl4 administration, TIMP-1 staining was significantly accentuated in groups I and III. IL-10 gene therapy (group II) attenuated this upregulation. (A) Normal control; (B) Group I; (C) Group II; (D) Group III. Arrows indicate TIMP-1-positive cells. Magnification, ×400 (inset, ×200).

Discussion

Animal models of hepatic fibrosis provide a means to study the cellular and molecular mediators of fibrosis in a serial manner during both progression and recovery. Several approaches to the induction of fibrosis have been described. Of these, CCl4 intoxication in rats and mice is probably the most widely studied[29]. In addition, the CCl4 model is the best characterized with respect to histological, biochemical, cellular, and molecular changes associated with the development of fibrosis[30,31]. CCl4 can be given intraperitoneally or by oral gavage; it induces hepatocyte necrosis and apoptosis with associated HSC activation and tissue fibrosis. With ongoing treatment CCl4 can be used to induce bridging hepatic fibrosis (4 weeks of twice-weekly treatment), cirrhosis (8 weeks of twice-weekly treatment) and advanced micronodular cirrhosis (12 weeks of twice-weekly treatment)[31].

IL-10 is a potent anti-inflammatory cytokine that inhibits the synthesis of pro-inflammatory cytokines[32]. IL-10 has been shown to downregulate the synthesis of collagen type I and TIMP in previous investigations[33,34]. It also plays an anti-fibrogenic role by decreasing the levels of pro-fibrogenic cytokines, including TGF-β1 and TNF-α[33]. In the present study, we demonstrated that electroporative IL-10 gene therapy provided an effective expression method for long-term use. This treatment reversed established liver fibrosis and reduced collagen synthesis in mice. IL-10 gene therapy also inhibited HSC activation after CCl4 administration. The fibrogenic gene (TGF-β1 and TNF-α) response attenuation may be responsible for the hepatoprotective effect of IL-10.

COX-2 is a key executor of uncontrolled inflammation[35]. Overexpression of COX-2 has been demonstrated in CCl4-induced liver fibrosis and post-viral human cirrhosis[36,37]. Further, COX-2 can contribute to hepatic carcinogenesis by increasing necroinflammatory activity, promoting proliferation, and enhancing angiogenesis[38,39]. Selective COX-2 blockers are known to reduce CCl4-induced liver fibrosis[36]. Hence, COX-2 may be a new therapeutic target for treatments for liver cirrhosis. IL-10 is known as the central regulator of COX-2[40]. Therefore, IL-10 gene therapy might have exerted its anti-hepatic fibrogenesis effect through COX-2 inactivation.

Cell adhesion molecules are known as prognostic markers of liver fibrosis[41]. Expression of ICAM-1 and VCAM-1 modulated by TNF-α are upregulated in alcoholic hepatitis, CCl4-induced liver injury, and nutritional fibrosis[4244]. A previous study showed that ICAM-1 and VCAM were upregulated in IL-10 knockout-colitis in mice[45]. In addition, IL-10 can attenuate ICAM-1 activation in cisplatin nephrotoxicity[46]. Therefore, cell adhesion molecule regulation may be involved in the anti-fibrotic effect of IL-10.

The imbalance between MMP and TIMP in the ECM contributes to the pathogenesis of liver fibrosis. Matrix metalloproteinases are a family of zinc-dependent proteases capable of degrading hepatic ECM, thereby playing a central role in tissue remodeling and repair after injury[47]; however, persistent overexpression of MMP may contribute to the pathogenesis of liver diseases. Inhibition of MMP-2 produced by activated stellate cells blocks lethal hepatitis and apoptosis induced by TNF-α[48]. Furthermore, MMP-2-deficient mice have decreased hepatocyte apoptosis and necrosis, and enhancsed survival in this model. Recent studies have also revealed a strong correlation between MMP-2 activity and severity of human liver disease[49]. MMP activity is regulated by the TIMP, which binds in a substrate- and tissue-specific manner to MMP, blocking their proteolytic activity[50]. Antibodies and antisense oligonucleotides directed at TIMP-1 attenuate rat liver fibrosis[50,51]. IL-10 is known to suppress MMP-2 and TIMP-1 expression in HSC during liver fibrosis[52]. In the present study, we demonstrated that IL-10 gene therapy attenuated MMP-2 and TIMP activation in the fibrotic liver. Therefore, its collagenolytic effect might be attributed to MMP and TIMP modulation.

In the present study, we demonstrated the anti-hepatic fibrogenic effect of IL-10 in mice. IL-10 gene therapy reversed established CCl4-induced liver fibrosis in mice through fibrogenic gene response attenuation. In conclusion, IL-10 gene therapy may be a new therapeutic modality for liver cirrhosis with potential clinical use.


Acknowledgement

We would like to thank Dr Xian-min MENG of Thomas Jefferson University, Philadelphia, PA, USA, for providing the human IL-10 expression plasmid (pCYIL-10 vector) used in this study.


References

  1. Kershenobich Stalnikowitz D, Weissbrod AB. Liver fibrosis and inflammation. A review. Ann Hepatol 2003;2:159-63.
  2. Pinzani M, Marra F. Cytokine receptors and signaling in hepatic stellate cells. Semin Liver Dis 2001;21:397-416.
  3. Carini R, Chiarpotto E, Biasi F, Leonarduzzi G, Comoglio A, Carpi C, et al. Relation between liver necrosis and intrahepatic cholestasis in rats poisoned with CCl4. Boll Soc Ital Biol Sper 1987;63:273-80.
  4. Recknagel RO, Glende EA Jr, Dolak JA, Waller RL. Mechanisms of carbon tetrachloride toxicity. Pharmacol Ther 1989;43:139-54.
  5. Schuppan D, Ruehl M, Somasundaram R, Hahn EG. Matrix as a modulator of hepatic fibrogenesis. Semin Liver Dis 2001;21:351-72.
  6. Giannelli G, Quaranta V, Antonaci S. Tissue remodelling in liver diseases. Histol Histopathol 2003;18:1267-74.
  7. Okazaki I, Watanabe T, Hozawa S, Arai M, Maruyama K. Molecular mechanism of the reversibility of hepatic fibrosis: with special reference to the role of matrix metalloproteinases. J Gastroenterol Hepatol 2000;15 Suppl:D26-32.
  8. Desmet VJ, Roskams T. Cirrhosis reversal: a duel between dogma and myth. J Hepatol 2004;40:860-7.
  9. McCrudden R, Iredale JP. Liver fibrosis, the hepatic stellate cell and tissue inhibitors of metalloproteinases. Histol Histopathol 2000;15:1159-68.
  10. Arthur MJ., Fibrogenesis II. Metalloproteinases and their inhibitors in liver fibrosis. Am J Physiol Gastrointest Liver Physiol 2000;279:G245-9.
  11. Gressner AM, Weiskirchen R, Breitkopf K, Dooley S. Roles of TGF-β in hepatic fibrosis. Front Biosci 2002;7:d793-807.
  12. Tilg H, Wilmer A, Vogel W, Herold M, Nolchen B, Judmaier G, et al. Serum levels of cytokines in chronic liver diseases. Gastroenterology 1992;103:264-74.
  13. Asadullah K, Sterry W, Volk HD. Interleukin-10 therapy: review of a new approach. Pharmacol Rev 2003;55:241-69.
  14. Zdanov A. Structural features of the interleukin-10 family of cytokines. Curr Pharm Des 2004;10:3873-84.
  15. Nelson DR, Lauwers GY, Lau JY, Davis GL. Interleukin 10 treatment reduces fibrosis in patients with chronic hepatitis C: a pilot trial of interferon nonresponders. Gastroenterology 2000;118:655-60.
  16. Zhang ZL, Shen SX, Lin B, Yu LY, Zhu LH, Wang WP, et al. Intramuscular injection of interleukin-10 plasmid DNA prevented autoimmune diabetes in mice. Acta Pharmacol Sin 2003;24:751-6.
  17. Zhang ZL, Lin B, Yu LY, Shen SX, Zhu LH, Wang WP, et al. Gene therapy of experimental autoimmune thyroiditis mice by in vivo administration of plasmid DNA coding for human inter-leukin-10. Acta Pharmacol Sin 2003;24:885-90.
  18. Lindsay JO, Ciesielski CJ, Scheinin T, Brennan FM, Hodgson HJ. Local delivery of adenoviral vectors encoding murine interleukin 10 induces colonic interleukin 10 production and is therapeutic for murine colitis. Gut 2003;52:981-7.
  19. Andersen SR, Lambrecht LJ, Swan SK, Cutler DL, Radwanski E, Affrime MB, et al. Disposition of recombinant human interleukin-10 in subjects with various degrees of renal function. J Clin Pharmacol 1999;39:1015-20.
  20. Meng X, Sawamura D, Tamai K, Hanada K, Ishida H, Hashimoto I. Keratinocyte gene therapy for systemic diseases. Circulating interleukin 10 released from gene-transferred keratinocytes inhibits contact hypersensitivity at distant areas of the skin. J Clin Invest 1998;101:1462-7.
  21. Yao HW, Li J, Chen JQ, Xu SY. Inhibitory effect of leflunomide on hepatic fibrosis induced by CCl4 in rats. Acta Pharmacol Sin 2004;25:915-20.
  22. Molnar MJ, Gilbert R, Lu Y, Liu AB, Guo A, Larochelle N, et al. Factors influencing the efficacy, longevity, and safety of electro-poration-assisted plasmid-based gene transfer into mouse muscles. Mol Ther 2004;10:447-55.
  23. Lehr HA, van der Loos CM, Teeling P, Gown AM. Complete chromogen separation and analysis in double immunohistochemical stains using Photoshop-based image analysis. J Histochem Cytochem 1999;47:119-26.
  24. Vergnes L, Phan J, Strauss M, Tafuri S, Reue K. Cholesterol and cholate components of an atherogenic diet induce distinct stages of hepatic inflammatory gene expression. J Biol Chem 2003;278:42774-84.
  25. Hung KS, Lee TH, Chou WY, Wu CL, Cho CL, Lu CN, et al. Interleukin-10 gene therapy reverses thioacetamide-induced liver fibrosis in mice. Biochem Biophys Res Commun 2005;336:324-31.
  26. Kossakowska AE, Edwards DR, Lee SS, Urbanski LS, Stabbler AL, Zhang CL, et al. Altered balance between matrix metallopro-teinases and their inhibitors in experimental biliary fibrosis. Am J Pathol 1998;153:1895-902.
  27. Wang CH, Chen YJ, Lee TH, Chen YS, Jawan B, Hung KS, et al. Protective effect of MDL28170 against thioacetamide-induced acute liver failure in mice. J Biomed Sci 2004;11:571-8.
  28. Wang CH, Jawan B, Lee TH, Hung KS, Chou WY, Lu CN, et al. Single injection of naked plasmid encoding α-melanocyte-stimulating hormone protects against thioacetamide-induced acute liver failure in mice. Biochem Biophys Res Commun 2004;322:153-61.
  29. Constandinou C, Henderson N, Iredale JP. Modeling liver fibrosis in rodents. Methods Mol Med 2005;117:237-50.
  30. Rojkind M, Greenwel P. Animal models of liver fibrosis. Adv Vet Sci Comp Med 1993;37:333-55.
  31. Di Vinicius I, Baptista AP, Barbosa AA, Andrade ZA. Morphological signs of cirrhosis regression. Experimental observations on carbon tetrachloride-induced liver cirrhosis of rats. Pathol Res Pract 2005;201:449-56.
  32. Pestka S, Krause CD, Sarkar D, Walter MR, Shi Y, Fisher PB. Interleukin-10 and related cytokines and receptors. Annu Rev Immunol 2004;22:929-79.
  33. Safadi R, Ohta M, Alvarez CE, Fiel MI, Bansal M, Mehal WZ, et al. Immune stimulation of hepatic fibrogenesis by CD8 cells and attenuation by transgenic interleukin-10 from hepatocytes. Gastroenterology 2004;127:870-82.
  34. Reitamo S, Remitz A, Tamai K, Uitto J. Interleukin-10 modulates type I collagen and matrix metalloprotease gene expression in cultured human skin fibroblasts. J Clin Invest 1994;94:2489-92.
  35. Claria J. Cyclooxygenase-2 biology. Curr Pharm Des 2003;9:2177-90.
  36. Planaguma A, Claria J, Miquel R, Lopez-Parra M, Titos E, Masferrer JL, et al. The selective cyclooxygenase-2 inhibitor SC-236 reduces liver fibrosis by mechanisms involving non-parenchymal cell apoptosis and PPARgamma activation. FASEB J 2005;19:1120-2.
  37. Mohammed NA, Abd El-Aleem SA, El-Hafiz HA, McMahon RF. Distribution of constitutive (COX-1) and inducible (COX-2) cyclooxygenase in postviral human liver cirrhosis: a possible role for COX-2 in the pathogenesis of liver cirrhosis. J Clin Pathol 2004;57:350-4.
  38. Sung YK, Hwang SY, Kim JO, Bae HI, Kim JC, Kim MK. The correlation between cyclooxygenase-2 expression and hepatocellular carcinogenesis. Mol Cells 2004;17:35-8.
  39. Bae SH, Jung ES, Park YM, Kim BS, Kim BK, Kim DG, Ryu WS. Expression of cyclooxygenase-2 (COX-2) in hepatocellular carcinoma and growth inhibition of hepatoma cell lines by a COX-2 inhibitor, NS-398. Clin Cancer Res 2001;7:1410-8.
  40. Berg DJ, Zhang J, Lauricella DM, Moore SA. IL-10 is a central regulator of cyclooxygenase-2 expression and prostaglandin production. J Immunol 2001;166:2674-80.
  41. Giron-Gonzalez JA, Martinez-Sierra C, Rodriguez-Ramos C, Rendon P, Macias MA, Fernandez-Gutierrez C, et al. Adhesion molecules as a prognostic marker of liver cirrhosis. Scand J Gastroenterol 2005;40:217-24.
  42. Burra P, Hubscher SG, Shaw J, Elias E, Adams DH. Is the intercellular adhesion molecule-1/leukocyte function associated antigen 1 pathway of leukocyte adhesion involved in the tissue damage of alcoholic hepatitis? Gut 1992;33:268-71.
  43. Simeonova PP, Gallucci RM, Hulderman T, Wilson R, Kommineni C, Rao M, et al. The role of tumor necrosis factor-alpha in liver toxicity, inflammation, and fibrosis induced by carbon tetrachloride. Toxicol Appl Pharmacol 2001;177:112-20.
  44. Ip E, Farrell G, Hall P, Robertson G, Leclercq I. Administration of the potent PPARα agonist, Wy-14,643, reverses nutritional fibrosis and steatohepatitis in mice. Hepatology 2004;39:1286-96.
  45. Kawachi S, Jennings S, Panes J, Cockrell A, Laroux FS, Gray L, et al. Cytokine and endothelial cell adhesion molecule expression in interleukin-10-deficient mice. Am J Physiol Gastrointest Liver Physiol 2000;278:G734-43.
  46. Deng J, Kohda Y, Chiao H, Wang Y, Hu X, Hewitt SM, et al. Interleukin-10 inhibits ischemic and cisplatin-induced acute renal injury. Kidney Int 2001;60:2118-28.
  47. Bode W, Fernandez-Catalan C, Tschesche H, Grams F, Nagase H, Maskos K. Structural properties of matrix metalloproteinases. Cell Mol Life Sci 1999;55:639-52.
  48. Wielockx B, Lannoy K, Shapiro SD, Itoh T, Itohara S, Vandekerckhove J, et al. Inhibition of matrix metallopro-teinases blocks lethal hepatitis and apoptosis induced by tumor necrosis factor and allows safe antitumor therapy. Nat Med 2001;7:1202-8.
  49. Chen TY, Hsieh YS, Yang CC, Wang CP, Yang SF, Cheng YW, et al. Relationship between matrix metalloproteinase-2 activity and cystatin C levels in patients with hepatic disease. Clin Biochem 2005;38:632-8.
  50. Parsons CJ, Bradford BU, Pan CQ, Cheung E, Schauer M, Knorr A, et al. Antifibrotic effects of a tissue inhibitor of metallopro-teinase-1 antibody on established liver fibrosis in rats. Hepatology 2004;40:1106-15.
  51. Nie QH, Cheng YQ, Xie YM, Zhou YX, Cao YZ. Inhibiting effect of antisense oligonucleotides phosphorthioate on gene expression of TIMP-1 in rat liver fibrosis. World J Gastroenterol 2001;7:363-9.
  52. Zheng WD, Zhang LJ, Shi MN, Chen ZX, Chen YX, Huang YH, et al. Expression of matrix metalloproteinase-2 and tissue inhibitor of metalloproteinase-1 in hepatic stellate cells during rat hepatic fibrosis and its intervention by IL-10. World J Gastroenterol 2005;11:1753-8.
Cite this article as: Chou Wy, Lu Cn, Lee Th, Wu Cl, Hung Ks, Concejero AM, Jawan B, Wang Ch. Electroporative interleukin-10 gene transfer ameliorates carbon tetrachloride-induced murine liver fibrosis by MMP and TIMP modulation1. Acta Pharmacologica Sinica 2006;27(4):469-476. doi: 10.1111/j.1745-7254.2006.00304.x