Hepatic stellate cells promote immunotolerance following orthotopic liver transplantation in rats via induction of T cell apoptosis and regulation of Th2/Th3‑like cell cytokine production

  • Authors:
    • Zhijun Jiang
    • Ying Chen
    • Xiaonin Feng
    • Jianwen Jiang
    • Tianxiang Chen
    • Haiyang Xie
    • Lin Zhou
    • Shusen Zheng
  • View Affiliations

  • Published online on: November 6, 2012     https://doi.org/10.3892/etm.2012.801
  • Pages: 165-169
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hepatic stellate cells (HSCs) have been demonstrated to have immunoinhibitory activity. The aim of this study was to investigate the role of HSCs in the development of immunotolerance following liver transplantation. A rat liver transplantation tolerance model [donor Lewis into recipient Dark Agouti (DA)] and rejection (donor DA into recipient Lewis) was established. On the 7th day following transplantation, the HSCs and T cells were isolated from the rats of either the tolerance or rejection group and cultured together. The apoptosis rate of the T cells was determined 24 h later by flow cytometry following staining with anti‑CD3 mAb and Annexin V‑FITC/PI. Additionally, the FasL expression of HSCs was determined by flow cytometry following staining with anti‑FasL mAb. The protein levels of IL‑2, TNF‑α, TGF‑β and IL‑10 in the supernatant collected from mixed lymphocyte reaction cultures of HSCs and T cells for 5 days were measured using ELISA assays. HSCs isolated from the tolerance group had a higher T‑cell apoptosis induction activity compared with those of the rejection group. The activity of the HSCs was partially reversed by FasL blocking mAb. Accordingly, the FasL expression level of HSCs in the tolerance group was revealed to be higher than that of the rejection group. Moreover, HSCs stimulated IL‑10 and TGF‑β production in the tolerance group. This study suggests that HSCs are involved in liver transplantation immune tolerance via the induction of T‑cell apoptosis partially mediated by the Fas/FasL pathway and the activation of Th2/Th3‑like cell cytokine production.

Introduction

Although organ transplantation has been successful for decades, graft rejection and immunosuppression (IS)-derived chronic toxicity remain as key problems which need to be overcome. However, in comparison with the majority of transplant recipients who have extreme difficulty achieving an IS-free state following transplantation, a significantly higher proportion of liver transplant recipients achieve clinical operational tolerance (1). Moreover, liver allografts are able to protect co-transplanted organs from rejection (24), suggesting that the liver, unlike other solid organs such as the kidney or heart, is an immunoprivileged organ. Notably, although liver allografts are the most spontaneously accepted transplantations in a number of species, hepatocyte allografts alone are acutely rejected (5,6), suggesting that liver non-parenchymal cells (NPCs), including resident dendritic cells (DCs), liver sinusoid endothelial cells (LSECs), Kupffer cells (KCs) and hepatic stellate cells (HSCs), are considerably involved in liver immunotolerance. Previous studies have indicated that NPCs mediate immunosuppresion via a variety of mechanisms, including the secretion of anti-inflammatory cytokines and the induction of T-cell apoptosis (7), However, the underlying mechanism is not yet completely understood.

HSCs (vitamin A-storing cells, lipocytes, interstitial cells, fat-storing cells and Ito cells), exist in the space between the hepatocytes and LSECs of the hepatic lobule and are well known for their functions of regulating retinoid homeostasis and participating in the pathogenesis of liver fibrogenesis. In addition, HSCs have been demonstrated to be antigen-presenting cells (APCs) and tolerogenic (8). Activated HSCs express negative co-stimulator B7-H1 which inhibits T-cell responses via the mediation of T-cell apoptosis (9). Previous studies have revealed that co-transplanted HSCs protect islet allografts from rejection and attenuate the severity of graft-versus-host disease (10,11), suggesting that the HSCs have immunosuppressive properties. The present study revealed that HSCs are involved in liver transplant immunotolerance via T-cell apoptosis partly mediated by the Fas/FasL pathway and the regulation of TGF-β and IL-10 production.

Materials and methods

Animals

Male Dark Agouti (DA) and Lewis rats, aged between 10 and 12 weeks and weighing 220–250 g (Vital River Laboratory Animal Technology Co., Ltd., Beijing, China) were maintained in a pathogen-free animal facility. The rats were allowed free access to tap water and food. The animal procedures were approved by the Institutional Animal Care Committee.

Orthotopic liver transplantation

Previous studies have shown that liver grafts are spontaneously accepted in Lewis to DA transplantations, while DA to Lewis liver allograft recipients suffer from severe rejection (12). The present study included a tolerance group (Lewis into DA, n=5) and rejection group (DA into Lewis, n=5). The liver transplantation was performed according to the ‘2 cuff technique’ of Kamada and Calne (13). Briefly, after anesthesia and systemic heparinization, the livers were removed from the donors and prepared in 4°C Ringer’s solution. The grafts were then implanted into the recipients by anastomosis of the suprahepatic vena cava using a continuous everting suture, reconstruction of the portal vein and infra-hepatic vena cava by the cuff technique and connection of bile duct by an end-to-end anastomosis over an indwelling stent without hepatic artery reconstruction. After surgery, the recipients were kept warm by lighting and had free access to food and water. Rats deaths within 5 days after transplantation were considered to be due to technical failures and hence excluded from the study.

Histological examination

Tissue specimens from the liver grafts were fixed in 4% formalin, embedded in paraffin and used for histological examination. The specimens were then sliced into 5-μm sections and stained with hematoxylin and eosin (H&E) for routine histological examination.

Isolation of HSCs

On the 7th day after transplantation, the recipient rats were sacrificed. A 0.5×1.0 cm liver tissue specimen was obtained from each liver graft for histological examination. HSCs were then extracted from the left liver tissue as previously described (1416). Briefly, the grafts underwent serial in situ perfusions with 70 ml 0.1% pronase at a flow rate of 10–15 ml/min for 7 min and 60 ml 0.05% collagenase IV (Sigma, St. Louis, MO, USA) at a flow rate of 10–15 ml/min for 20 min. The liver tissue was then digested in 50 ml buffer solution containing collagenase IV, pronase and DNase (Sigma), followed by density gradient centrifugation and 11% Nycodenz (Axis-Shield PoC, Oslo, Norway) gradient centrifugation. The harvested HSCs were resuspended in high glucose Dulbecco’s modified Eagle’s medium (DMEM; Gibco-BRL, Grand Island, NY, USA) containing 20% fetal calf serum (FCS). The viability of HSCs was >90% as determined using trypan blue exclusion and the purity of HSCs ranged from 90 to 95% as determined by desmin immunostaining. The typical light microscopic appearance of a lipid droplet was as described previously (17). The HSCs were cultured in DMEM containing 20% FCS for 7 days for further study.

Preparation of T cells

The spleens of the recipient rats were removed on the 7th day after transplantation and single spleen cell (SC) suspensions were prepared. Following lysis of red blood cells and density gradient centrifugation, T cells were isolated and purified using an adherence culture in DMEM containing 10% FCS and a nylon wool column.

FasL expression in HSCs

HSCs (2.5×104) were incubated with mitomycin C (10 μg/ml) for 30 min at 37°C in a 5% CO2-humidified air atmosphere. Then, HSCs were stained with Hamster monoclonal anti-rat FasL-specific IgG (eBioscience, San Diego, CA, USA) followed by a second fluorescein-labeled antibody (BD Pharmingen, San Diego, CA, USA). The FasL expression levels of the HSCs were determined by flow cytometry according to the manufacturer’s instructions.

Flow cytometry analysis for alloreactive T-cell apoptosis

After incubating with mitomycin C (10 μg/ml) for 30 min, the HSCs (2.5×104) were resuspended in 100 μl DMEM containing 10% FCS and then co-cultured with 100 μl T cells (5×105) in the presence or absence of FasL blocking mAb for 24 h. All cultures were incubated at 37°C in a 5% CO2-humidified air atmosphere. The nonadherent cells were isolated and the apoptotic T cells stained with anti-CD3 mAb (eBioscience) and Annexin V-FITC/PI (BD Pharmingen) were determined by flow cytometry according to the manufacturer’s instructions.

Cytokine quantitation

A two-way mixed lymphocyte reaction (MLR) was performed and 2.5×105 Lewis and 2.5×105 DA SCs were co-cultured in 96-well plates (200 μl) in DMEM containing 10% FCS, 100 U/ml penicillin and 100 mg/ml streptomycin, in the presence of 2.5×104 HSCs. This co-culture was incubated at 37°C in a 5% CO2-humidified air atmosphere for 5 days. The IL-2, IL-10, TNF-α and TGF-β levels in the supernatant of this co-culture were quantified using the respective ELISA kits (Biosource International Inc, Camarillo, CA, USA). A standard curve using recombinant cytokine was generated for each assay.

Statistical analysis

The data are presented as the mean ± standard deviation (SD). The statistical significance of the parametric data was determined using the Student’s t-test. P<0.05 was considered to indicate a statistically significant difference.

Results

Histological characteristics of the liver grafts

Histological analysis confirmed that the liver grafts of the rejection group underwent serious acute rejection, while those of the tolerance group did not. In the rejection group, the H&E staining results showed severe acute rejection, which was characterized by extensive T-cell infiltration of the portal tracts; bile duct damage; vacuolar degeneration, karyopycnosis and even necrosis of hepatocytes. Meanwhile, the tolerance group exhibited only minimal T-cell infiltration without portal tract involvement and the hepatic parenchyma exhibited no significant damage (Fig. 1).

HSCs induce T-cell apoptosis through the Fas/FasL pathway

As T-cell apoptosis is a well-established mechanism of liver immunotolerance (18), we hypothesized that HSCs may contribute to liver immunotolerance by inducing T-cell apoptosis. The results indicated that the HSCs of the tolerance group had significantly higher FasL expression than the rejection group (Fig. 2). Furthermore, the results also showed that the HSCs of the tolerance group were able to markedly enhance the apoptosis of T cells, suggesting that the immunosuppressive effect of HSCs may induce T-cell apoptosis (Fig. 3). The Fas/FasL is a well-known pathway of apoptosis (19). To determine whether Fas/FasL was critical in HSC-induced T-cell apoptosis, HSCs were co-cultured with T cells in the presence or absence of FasL blocking mAb. FasL blocking mAb partially but significantly inhibited T-cell apoptosis in the tolerance group (P<0.05) and had little effect in the rejection group (P>0.05; Figs. 4 and 5).

HSCs promote Th2/Th3-like cell cytokine production in the tolerance group

The IL-2, TNF-α, TGF-β and IL-10 cytokine levels in the supernatant of the MLR cultures in the presence of HSCs were measured using ELISA assays. The results indicated that there were no significant differences in the Th1 cytokine (IL-2 and TNF-α) levels between the tolerance and rejection groups (P>0.05). However, in the tolerance group, the Th2-like cell cytokine IL-10 and Th3-like cell cytokine TGF-β levels were markedly increased (P<0.05; Fig. 6).

Discussion

Unlike other solid organs, liver allografts may be spontaneously accepted without IS in a number of species, including humans, demonstrating that the liver is an immunoprivileged organ (1,2022). Moreover, an accumulating amount of evidence suggests that NPCs, such as DCs, KCs and LSECs, are critical in liver transplant immunotolerance (2325). HSCs are a type of NPC, well known for their key role in liver fibrogenesis. Additionally, HSCs have been demonstrated to be immunoregulating (810).

In the present study, the activated HSCs from the tolerance group had an increased T-cell apoptosis-inducing activity, indicating that activated HSCs have an immune suppressive function, since induced T-cell apoptosis is a significant mechanism in the development of immune tolerance (18). The present data also showed that in the tolerance group, the expression of FasL by HSCs was significantly higher than in the rejection group. Moreover, FasL blocking mAb partially but significantly reversed HSC-induced T-cell apoptosis in the tolerance group but not in the rejection group, suggesting that the Fas/FasL pathway is associated with HSC-induced T-cell apoptosis. It is generally accepted that Fas and FasL interactions are key to cell apoptosis and maintain the immunoprivilege (19). Moreover, other studies have revealed that the Fas system is involved in liver transplant immune regulation. Specifically, FasL expressed by infiltrating cells induces liver cell apoptosis during acute rejection following liver transplantation. However, the increased expression of FasL in liver allografts results in immunotolerance by combining with Fas expressed by the infiltrating lymphocytes (26,27). This phenomenon may be explained by the expression of FasL gradually switching from infiltrating cells to hepatocytes. Sun et al observed that KCs in the liver downregulate the T-cell response via the Fas/FasL pathway following liver transplantation. The authors results indicated that the Fas/FasL pathway was involved in the immunotolerance of liver transplantation (24). However, FasL may not be the only molecule involved in the immunosuppressive effect of HSCs, since the blocking of FasL only partially reversed HSC-induced T-cell apoptosis. Studies have further demonstrated that the upregulation of B7-H1 suppresses T-cell proliferation, promotes T-cell apoptosis and induces the production of various cytokines by combining with programmed death-1 (PD-1) B and T lymphocyte attenuator. Therefore, B7-H1 is considerably involved in peripheral immune tolerance and tumor immune evasion (28,29). B7-H1 negatively regulates the immune system and inhibits T-cell activity, mainly at the effect phase since B7-H1 receptor PD-1 is inducibly expressed on activated T cells (3032). Deficiencies of B7-H1 lead to the accumulation of CD8+ T cells in the liver, suggesting a role for B7-H1 in the regulation of T-cell homeostasis (33). A study by Yu et al showed that quiescent HSCs express very low levels of B7-H1, while B7-H1 expression in HSCs may be notably increased by various stimuli and the inhibition of B7-H1 may partially reduce HSC-induced T-cell apoptosis (9). Therefore, we propose that the B7-H1 and Fas/FasL pathways are involved in HSC-induced immune suppression.

The present study also showed that there were higher IL-10 and TGF-β levels in the supernatant of the MLR cultures of HSC and T cells from the tolerance group. We hypothesize that HSCs may drive the T-cell subset differentiation of Th2/Th3 cells or activate the production of inhibitory cytokines, such as IL-10 and/or TGF-β, by HSCs. Th1-like cell cytokines mediate cellular immunity and enhance rejection, while Th2-like cell cytokines downregulate the activity of Th1-like cells and cytotoxic T lymphocytes (CTL) and attenuate post-transplantation rejection. It has been reported that IL-10 and TGF-β contributed to liver transplant immunotolerance (34,35). Studies have also demonstrated that KCs and LSECs negatively regulate the immune response by secreting TGF-β (35). Therefore, HSCs may regulate the immune response following liver transplantation by regulating Th2/Th3-like cell cytokine production.

In conclusion, the present study revealed that HSCs contribute to liver transplant immunotolerance by inducing T-cell apoptosis and stimulating Th2/Th3-like cell cytokine production. This immunosuppressive activity of HSCs provides a supplementary mechanism for the development of immunotolerance following liver transplantation.

Acknowledgements

This study was supported by the Foundation for Innovative Research Groups of the National Natural Science Foundation of China (No.81121002) and Zhejiang Provincial Natural Science Foundation of China (No.Y 2100498).

References

1 

Orlando G, Soker S and Wood K: Operational tolerance after liver transplantation. J Hepatol. 50:1247–1257. 2009. View Article : Google Scholar

2 

Zhong R, He G, Sakai Y, Li XC, Garcia B, Wall W, Duff J, Stiller C and Grant D: Combined small bowel and liver transplantation in the rat: possible role of the liver in preventing intestinal allograft rejection. Transplantation. 52:550–552. 1991. View Article : Google Scholar : PubMed/NCBI

3 

Wang C, Sun J, Wang L, Li L, Horvat M and Sheil R: Combined liver and pancreas transplantation induces pancreas allograft tolerance. Transplant Proc. 29:1145–1146. 1997. View Article : Google Scholar : PubMed/NCBI

4 

Sarnacki S, Révillon Y, Cerf-Bensussan N, Calise D, Goulet O and Brousse N: Long-term small-bowel graft survival induced by a spontaneously tolerated liver allograft in inbred rat strains. Transplantation. 54:383–385. 1992. View Article : Google Scholar : PubMed/NCBI

5 

Bumgardner GL and Orosz CG: Unusual patterns of alloimmunity evoked by allogeneic liver parenchymal cells. Immunol Rev. 174:260–279. 2000. View Article : Google Scholar : PubMed/NCBI

6 

Bumgardner GL, Heininger M, Li J, Xia D, Parker-Thornburg J, Ferguson RM and Orosz CG: A functional model of hepatocyte transplantation for in vivo immunologic studies. Transplantation. 65:53–61. 1998. View Article : Google Scholar : PubMed/NCBI

7 

Tiegs G and Lohse AW: Immune tolerance: what is unique about the liver. J Autoimmun. 34:1–6. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Winau F, Hegasy G, Weiskirchen R, Weber S, Cassan C, Sieling PA, Modlin RL, Liblau RS, Gressner AM and Kaufmann SH: Ito cells are liver-resident antigen-presenting cells for activating T cell responses. Immunity. 26:117–129. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Yu MC, Chen CH, Liang X, Wang L, Gandhi CR, Fung JJ, Lu L and Qian S: Inhibition of T cell responses by hepatic stellate cells via B7-H1-mediated T-cell apoptosis in mice. Hepatology. 40:1312–1321. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Chen CH, Kuo LM, Chang Y, Wu W, Goldbach C, Ross MA, Stolz DB, Chen L, Fung JJ, Lu L and Qian S: In vivo immune modulatory activity of hepatic stellate cells in mice. Hepatology. 44:1171–1181. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Chen CH, Shu KH, Su YH, Tang KY, Cheng CH, Wu MJ, Yu TM, Chuang YW and Hu C: Cotransplantation of hepatic stellate cells attenuates the severity of graft-versus-host disease. Transplant Proc. 42:971–975. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Kamada N: The immunology of experimental liver transplantation in the rat. Immunology. 55:369–389. 1985.PubMed/NCBI

13 

Kamada N and Calne RY: Orthotopic liver transplantation in the rat. Technique using cuff for portal vein anastomosis and biliary drainage. Transplantation. 28:47–50. 1979. View Article : Google Scholar : PubMed/NCBI

14 

Niki T, Pekny M, Hellemans K, Bleser PD, Berg KV, Vaeyens F, Quartier E, Schuit F and Geerts A: Class VI intermediate filament protein nestin is induced during activation of rat hepatic stellate cells. Hepatology. 29:520–527. 1999. View Article : Google Scholar : PubMed/NCBI

15 

Iredale JP, Benyon RC, Pickering J, McCullen M, Northrop M, Pawley S, Hovell C and Arthur MJ: Mechanisms of spontaneous resolution of rat liver fibrosis hepatic stellate cell apoptosis and reduced hepatic expression of metalloproteinase inhibitors. J Clin Invest. 102:538–549. 1998. View Article : Google Scholar

16 

Lang A, Schoonhoven R, Tuvia S, Brenner DA and Rippe RA: Nuclear factor kappaB in proliferation, activation, and apoptosis in rat hepatic stellate cells. J Hepatol. 33:49–58. 2000. View Article : Google Scholar : PubMed/NCBI

17 

Liu C, Gaça MD, Swenson ES, Vellucci VF, Reiss M and Wells RG: Smads 2 and 3 are differentially activated by transforming growth factor-beta (TGF-beta) in quiescent and activated hepatic stellate cells. Constitutive nuclear localization of Smads in activated cells is TGF-beta-independent. J Biol Chem. 278:11721–11728. 2003. View Article : Google Scholar

18 

Qian S, Lu L, Fu F, Li Y, Li W, Starzl TE, Fung JJ and Thomson AW: Apoptosis within spontaneonsly accepted mouse liver allografts: evidence for deletion of cytotoxic T cells and implications for tolerance induction. J Immunol. 158:4654–4661. 1997.

19 

Griffith TS, Brunner T, Fletcher SM, Green DR and Ferguson TA: Fas ligand-induced apoptosis as a mechanism of immune privilege. Science. 270:1189–1192. 1995. View Article : Google Scholar : PubMed/NCBI

20 

Garnier H, Clot JP, Bertrand M, Camplez P, Kunlin A, Gorin JP, Le Goaziou F, Lévy R and Cordier G: Liver transplantation in the pig: surgical approach. C R Acad Sci Hebd Seances Acad Sci D. 260:5621–5623. 1965.(In French).

21 

Kamada N, Brons G and Davies HS: Fully allogeneic liver grafting in rats induces a state of systemic nonreactivity to donor transplantation antigens. Transplantation. 29:429–431. 1980. View Article : Google Scholar

22 

Qian S, Demetris AJ, Murase N, Rao AS, Fung JJ and Starzl TE: Murine liver allograft transplantation: tolerance and donor cell chimerism. Hepatology. 19:916–924. 1994. View Article : Google Scholar : PubMed/NCBI

23 

Fairchild PJ and Waldmann H: Dendritic cells and prospects for transplantation tolerance. Curr Opin Immunol. 12:528–535. 2000. View Article : Google Scholar : PubMed/NCBI

24 

Sun Z, Wada T, Maemura K, Uchikura K, Hoshino S, Diehl AM and Klein AS: Hepatic allograft-derived Kupfer cells regulate T cell response in rats. Liver Transpl. 9:489–497. 2003. View Article : Google Scholar : PubMed/NCBI

25 

Limmer A and Knolle PA: Liver sinusoidal endothelial cells: a new type of organ-resident antigen-presenting cell. Arch Immunol Ther Exp (Warsz). 49(Suppl 1): S7–S11. 2001.PubMed/NCBI

26 

Pan TL, Goto S, Lin YC, Lord R, Chiang KC, Lai CY, Chen YS, Eng HL, Cheng YF, Tatsuma T, Kitano S, Lin CL and Chen CL: The fas and fas 1igand pathways in liver allograft tolerance. Clin Exp Immunol. 118:180–187. 1999. View Article : Google Scholar : PubMed/NCBI

27 

Rivero M, Crespo J, Mayorga M, et al: Involvement of the Fas system in liver allograft rejection. Am J Gastroenterol. 97:1501–1506. 2002. View Article : Google Scholar : PubMed/NCBI

28 

Selenko-Gebauer N, Majdic O, Szekeres A, Höfler G, Guthann E, Korthäuer U, Zlabinger G, Steinberger P, Pickl WF, Stockinger H, Knapp W and Stöckl J: B7-H1 (Programmed death-1 ligand) on dendritic cells is involved in the induction and maintenance of T cell anergy. J Immunol. 170:3637–3644. 2003. View Article : Google Scholar : PubMed/NCBI

29 

Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, Roche PC, Lu J, Zhu G, Tamada K, Lennon VA, Celis E and Chen L: Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 8:793–800. 2002. View Article : Google Scholar : PubMed/NCBI

30 

Lu L, Qian S, Hershberger PA, Rudert WA, Lynch DH and Thomson AW: Fas ligand (CD95L) and B7 expression on dendritic cells provide counterregulatory signals for T cell survival and proliferation. J Immunol. 158:5676–5684. 1997.PubMed/NCBI

31 

Savage CO, Hughes CC, Pepinsky RB, Wallner BP, Freedman AS and Pober JS: Endothelial cell lymphocyte function-associated antigen-3 and an unidentified ligand act in concert to provide costimulation to human peripheral blood CD4+ T cells. Cell Immunol. 137:150–163. 1991.PubMed/NCBI

32 

Chen L: B7-H1 connection of innate and adoptive immunity against tumor dormancy. Blood. 105:2242–2243. 2005. View Article : Google Scholar

33 

Subudhi SK, Alegre ML and Fu YX: The balance of immune responses: costimulation verse coinhibition. J Mol Med (Berl). 83:193–202. 2005. View Article : Google Scholar : PubMed/NCBI

34 

Fernandes H, Koneru B, Fernandes N, Hameed M, Cohen MC, Raveche E and Cohen S: Investigation of promoter polymorphisms in the tumor necrosis factor-alpha and interleukin-10 gene in liver transplant patients. Transplantation. 73:1886–1891. 2002. View Article : Google Scholar : PubMed/NCBI

35 

Karrar A, Broomé U, Uzunel M, Qureshi AR and Sumitran-Holgersson S: Human liver sinusoidal endothelial cells induce apoptosis in activated T cells: a role in tolerance induction. Gut. 56:243–252. 2007. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January 2013
Volume 5 Issue 1

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Jiang Z, Chen Y, Feng X, Jiang J, Chen T, Xie H, Zhou L and Zheng S: Hepatic stellate cells promote immunotolerance following orthotopic liver transplantation in rats via induction of T cell apoptosis and regulation of Th2/Th3‑like cell cytokine production. Exp Ther Med 5: 165-169, 2013
APA
Jiang, Z., Chen, Y., Feng, X., Jiang, J., Chen, T., Xie, H. ... Zheng, S. (2013). Hepatic stellate cells promote immunotolerance following orthotopic liver transplantation in rats via induction of T cell apoptosis and regulation of Th2/Th3‑like cell cytokine production. Experimental and Therapeutic Medicine, 5, 165-169. https://doi.org/10.3892/etm.2012.801
MLA
Jiang, Z., Chen, Y., Feng, X., Jiang, J., Chen, T., Xie, H., Zhou, L., Zheng, S."Hepatic stellate cells promote immunotolerance following orthotopic liver transplantation in rats via induction of T cell apoptosis and regulation of Th2/Th3‑like cell cytokine production". Experimental and Therapeutic Medicine 5.1 (2013): 165-169.
Chicago
Jiang, Z., Chen, Y., Feng, X., Jiang, J., Chen, T., Xie, H., Zhou, L., Zheng, S."Hepatic stellate cells promote immunotolerance following orthotopic liver transplantation in rats via induction of T cell apoptosis and regulation of Th2/Th3‑like cell cytokine production". Experimental and Therapeutic Medicine 5, no. 1 (2013): 165-169. https://doi.org/10.3892/etm.2012.801