Review Open Access
Copyright ©2012 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastroenterol. Jan 14, 2012; 18(2): 119-125
Published online Jan 14, 2012. doi: 10.3748/wjg.v18.i2.119
Immune mechanisms of Concanavalin A model of autoimmune hepatitis
Hai-Xia Wang, College of Sciences, Anhui Science and Technology University, Fengyang 233100, Anhui Province, China
Man Liu, Shun-Yan Weng, Jing-Jing Li, Chao Xie, Hong-Lin He, Wen Guan, Yun-Sheng Yuan, Shanghai Municipality Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
Jin Gao, Laboratory of Regeneration, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
Author contributions: Wang HX and Liu M were the first authors; Wang HX and Liu M wrote the paper; Weng SY, Li JJ, Xie C, He HL, Guan W and Yuan YS contributed equally to this paper; Weng SY was in charge of revision; Li JJ and He HL contributed new drawing software and made the picture perfect; Xie C, Guan W and Yuan YS made a second revision; and Gao J brought up the idea and organized the manuscript.
Supported by Program for Excellent Talents of Anhui Province, No. 2006JQ1196
Correspondence to: Jin Gao, PhD, Laboratory of Regeneration, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Min Hang District, Shanghai 200240, China. g_jin@sjtu.edu.cn
Telephone: +86-21-34205769 Fax: +86-21-34204760
Received: April 20, 2011
Revised: July 6, 2011
Accepted: July 13, 2011
Published online: January 14, 2012

Abstract

As a chronic inflammatory disease of the liver, the pa-thogenic mechanisms of autoimmune hepatitis (AIH) have not yet been elucidated, with prognosis and diagnosis remaining unsatisfied. Currently the only viable treatments of AIH are immunosuppressant application and liver transplantation. It is considered that lack of good animal AIH models is the main reason for the shortage of a simple and efficient cure. The Concanavalin A (Con A) model is a typical and well established model for investigating T-cell and macrophage dependent liver injury in mice, which closely mimics the pathogenesis mechanisms and pathological changes of patients, and is regarded as the best experimental model for AIH research so far. In this paper we elucidated the pathogenic mechanisms of AIH and the evolution of relative animal models. We go on to further focus on Con A-induced liver injury from the point of immunological mechanisms and the change of cytokine levels. Finally, we manifested the clinical significance of the AIH animal models and the challenges they would meet during their future development.

Key Words: Autoimmune hepatitis, Animal models, Concanavalin A



INTRODUCTION

Autoimmune hepatitis (AIH) is a chronic inflammatory disease of the liver, characterized by a loss of self-tolerance leading to the appearance of autoantibodies, pathological changes and dysfunctions (the detailed pathogenic mechanisms of which still remain vague). According to different antibodies profiles, AIH is classified into three categories: AIH type 1 is characterized by the presence of antibodies to nuclear antigens (ANA) and/or anti-smooth muscle antigen (SMA) antibodies; AIH type 2 is characterized by anti-liver kidney microsomal (LKM)-1 and low level of LKM-3 antibodies (with or without ANA or SMA antibodies); AIH type 3 is characterized by autoantibodies against soluble liver antigen/liver pancreas (with or without ANA or SMA antibodies)[1].

Around the world, the incidence of AIH is 0.1-1.9 cases out of 100 000 persons per year, which is not very high[2]. However, the prevalence of autoimmune hepatitis in Europe is in the range of 11.6-16.9 cases per 100 000 persons[2], and in the United States, the proportion of hepatitis among patients with liver cancer is about 11%[3]. Incidence is also different between men and women. It was reported that women are more vulnerable to AIH[2,4,5].

Unfortunately, we do not have any better choice of medicines other than immunosuppressants, which can be classified into four generations[6,7]. In the 1950s, the first generation immunosuppressants were limited to azathioprine and steroids, which were enriched by polyclonal anti-lymphocyte and anti-thymocyte globulins in the 1960s[6]. For this generation, 70%-80% patients might relapse after withdrawal of treatment[8]. More seriously, they have many side effects[9]. Corticosteroids, Tacrolimus and Cyclosporine are typical of the second generation[6]. In the early 1990s a broad range of third-generation immunosuppressants emerged[6], most of which are monoclonal anti-lymphocyte and anti-thymocyte globulins followed by the fourth generation, such as the IL-2 monoclonal antibody with its highly specific sites of action[7,10]. The second and the third generation immunosuppressants are in most cases successfully used for treatment of AIH[11,12]. But long term applications of these immunosuppressive drugs carries serious risks[13] and sustained remission[9], even at low doses. Non-system steroids may be the best candidates[14]. Patients with liver failure or fulminant presentation who fail to improve under immunosuppressive therapy should be considered as candidates for liver transplantation. Without treatment, nearly 50% of patients with severe autoimmune hepatitis die in approximately 5 years[15]. Taking this into consideration, it is significantly important to develop new specific drugs. Animal models are the basis of drug discovery and development. Up to the time of writing, there are still no universal animal models of AIH which can be used as pathogenic models as well as therapeutic ones.

As the most important AIH research model, the Con A animal model plays a key role in AIH drug development. In this article we attempt to review the evolution of the Con A animal model of AIH, to sum up the mechanisms of Con A-induced liver injury, and to illustrate its statue in AIH drug development. Furthermore, the future challenges of the animal model are also discussed.

EVOLUTION OF AIH MODEL

AIH models have evolved from crude liver homogenates and adjuvants to the genetic engineering level, which can be classified into five phases[16]. The first phase was in 1972 when Buschenfelde et al[17] induced chronic active hepatitis in rabbits immunized with human liver proteins combined with complete Freund’s adjuvant. This work built a solid foundation for AIH models. The second phase began in 1983, when Mihas et al[18] established transient hepatitis in mice by immunization with syngeneic liver proteins together with the polysaccharide of Klebsiella pneumoniae. In the third phase, taking place from 1987 to 1990, many scientists used inbred or neonatal thymectomy mice to establish the T-cell reactive AIH model. They induced transient hepatitis by immunizing C57BL/6 mice with the supernatant of liver syngeneic liver homogenates with complete Freund’s adjuvant and used adoptive transfer technology to study the roles of T-cell, which allowed studies of the pathogenesis of AIH[19]. The fourth phase, from 1992 to 2003, had endotoxin and plant lectin-induced hepatitis models receive extensive attention. Three types of inducers were wildly used during this period: Con A[20], D-galactosamine (GalN) with low dosage of lipopolysaccharides (LPS)[21], and high dosage of LPS[22]. In the fifth phase, from 2002 to 2008, the application of genetic engineering technology accelerated the development of AIH model[23]. From one aspect, gene knockout and transgenic animals facilitated the study of the functions of certain genes[24]. From the other, production of designated antibodies using genetic engineering methods made it possible for scientists to get specific types of autoantibodies[25], and also made it possible for the Con A models to mimic a specific subtype of AIH. Significantly, the production of designated autoantibodies is based on known antigens. Scientists have now clarified the antigens to the following autoantibodies: the antigen to LKM-1 is cytochrome P450 2D6[26,27], the antigen to LKM-2 is cytochrome P450 2C9[1], the antigens to Liver Microsomal are cytochrome P450 1A2 and cytochrome P450 2A6[1]. The animal models of type 2 AIH[28] have been reported, but obviously type I animal models have more clinical significance than type II[2]. As is widely known, it is difficult to find the antigen of autoantibodies, which is the limitation of the gene engineering AIH model. The features and parameters of the three models are listed in Table 1[29].

Table 1 The features of the autoimmune hepatitis model induced by endotoxins and plant lectins.
Con A[29]GalN/LPS[29]LPS[29]
AnimalBALB/c-miceBALB/c-miceBALB/c-mice
(6-8 wk)(6-8 wk)(6-8 wk)
InducerCon AGalN/LPSLPS
Dosage20 mg/kgLPS: 5 μg/kg10 mg/kg
GalN: 700 mg/kg
Application methodTail veinSubcutaneousSubcutaneous
Transaminase level (max)8 h8 hNo significant change

From the information in Table 1, it is obvious that the Con A-induced hepatitis model possesses more advantages than the other two. Firstly, the Con A model includes only one inducer, making it easier to be established compared with the GalN/LPS model. Secondly, there is no significant change of the level of transaminase, which is considered a valid index of the severity of liver injury, in the LPS model, while such change is remarkable in the Con A model. Thirdly, in the Con A model, the serum level of many cytokines relevant to inflammation change dramatically, which is favorable for the study of the pathogenic mechanisms of AIH[29]. Furthermore, besides AIH, Con A animal models with different parameters are adaptable to many clinical diseases, such as fulminant hepatitis[30], virus hepatitis[31], hepatotoxin[32,33] and alcoholic liver diseases[34]. In summary, Con A AIH model is easy, convenient, inexpensive and repeatable, as well as a T-cell activated model and could greatly facilitate the study of the mechanisms of AIH-induced liver injury.

IMMUNOLOGICAL MECHANISMS OF CON A-INDUCED LIVER INJURY

Con A is one kind of lectin, which is purified from Canavalia brasiliensis[35]. Tiegs et al[20] injected Con A, Succinyl Con A with no agglutination activity, and Vicia faba lectin with strong agglutination activity to nuclear magnetic resonance imaging mice via tail vein, respectively. The results showed that only Con A could induce liver injury, which indicated that the in vitro agglutination activity of this lectin does not correlate with its hepatotoxic potential in vivo. They also studied the correlation between the hepatotoxic potential of Con A and its sugar-binding site[20]. Con A has specific sugar-binding sites, whose ligands are α-D-mannoside, methyl α-D-mannopyranoside, α-D-glucose, and methyl-α-D-glucose[36]. They co-administrated Con A with α-D-mannoside or methyl α-D-mannopyranoside to mice, which prevented the induction of hepatic injury by the lectin[20]. This suggested that free sugar-binding sites are indispensible for the induction of liver injury by lectin. Sato et al[37] also confirmed that Con A/glycogen multilayer films can be decomposed by exposing them to sugar solutions (D-glucose, D-mannose, methyl-alpha-D-glucose and methyl-alpha-D-mannose), as a result of the displacement of sugar residues of glycogen from the binding sites of Con A by the free sugar added in the solution. This suggested that sugar-binding sites are prerequisites of activated Con A. But among Con A, Succinyl Con A and Vicia faba lectin, which have the same sugar-binding site, only Con A can lead to high level of transaminase[20]. These two results indicated that the hepatotoxic potential of Con A is not determined by its agglutination activity or sugar-binding site. Other mechanisms may exist.

The mechanisms of the Con A model have interested many scientists. Previews papers describe that the aminotransferase of mice in thymus[38] and CD4[39] neutralized groups decreased significantly compared with the control group, while the CD8 neutralized group show no significant change. What is more, after injection of Con A, the blood level of interleukin 2 (IL-2), IL-4 and interferon gamma (IFN-γ) all increased dramatically[40]. This suggested that the CD4+ T helper (Th) cell was involved in the liver injury[40]. It is reported that CD4+-positive Th cells recognize the Con A-modified major histocompatibility complex (MHC) structures of macrophages and become activated, followed by an inflammation reaction and the release of IL-1 and IL-2 to the blood[41]. In the experiment of CD8 neutralization, there was a minor decrease of the transaminase level, which suggested that the target cell lysis by cytotoxic CD8+ T lymphocyte (CTL) also contributes to liver injury, but not as the major factor. In conclusion, the main mechanism of the Con A model is that Th cell activation increases the relevant cytokine level, which leads to liver injury. Meanwhile, the CTL mediated target cell lysis may be the secondary mechanism.

In the liver, lymphocytes, sinusoid endothelial cells (SECs), Kupffer cells (KCs) and stellate cells are all involved in the immune response[42]. Lymphocytes can be classified into two groups, exogenous and endogenous[43]. Exogenous lymphocytes originate from the thymus[44], bone marrow[44,45], intestinal tract[46], spleen[47] and lymph gland[48], and enter the liver through circulation. Endogenous lymphocytes are enriched in the portal area of the liver, which count for 25% of non-parenchyma cells in the liver[49]. The endogenous lymphocytes are mainly T cells, while B cells only count for 5% of them. This is why lymphocyte infiltration is mainly focused in the portal area[50].

For a long time, there have been debates about whe-ther KCs or SECs plays a major role in immunological liver injury[51-53]. Knolle et al[52] established the spontaneous and LPS activated cell model, and found that SECs and KCs both secreted IL-1 and IL-6, which suggested that SECs are also key cells in liver injury. It has been found that fifteen minutes after intravenous injection of Con A, Con A binds to SECs first; 4 h later, Con A begins to bind to the KCs[52]. Using Scanning Electron micrograph, it is clearly seen that 4 h after intravenous injection of Con A, blood cell endothelium attaches to the SECs first[52]. Then lymphocytes or neutrophils are trafficked into the hepatocytes, leading to inflammation[52]. We can conclude that SECs and KCs are both important, but they play their roles in the different phases. After injection, Con A binds to the mannose gland in the SECs surface first, leading to the breakdown of the SECs membrane, bleb formation and cytoplasm disappearance[30]. SECs detachment facilitates the binding of Con A to the KCs. CD4+ Th cells recognize the MHC class II and T cell receptor of KCs modified by Con A and are then activated[30]. Such liver injury is mainly mediated by T helper cells, including Th1 and Th2 cells. Figure 1 depicted the mechanisms of T cell activated liver injury.

Figure 1
Figure 1 Mechanisms of Concanavalin A induced T cell activated liver injury. Con A: Concanavalin A; KC: Kupffer cell; SEC: Sinusoid endothelial cell; MHC: Major histocompatibility complex.
CHANGES IN THE EXPRESSION LEVELS OF RELEVANT CYTOKINES

Some major cytokines involved in the Con A-induced liver injury are IFN-γ[54-55], IL-2[55], IL-4[56], IL-656] and tumor necrosis factor α (TNF-α)[56], of which TNF-α and IFN-γ are the major ones.

Figure 2 shows the time when different cytokines reach their peak level in the plasma and liver. In the plasma, TNF-α and IL-10[29,57] first reach their peak level after 1 h, followed by IL-4 after 2 h. IFN-γ, IL-2 and IL-6[29,57] reach their peak after 3 h, followed by IL-12. However, in the liver, TNF-α, IFN-γ, IL-4[29,57] reach their peak level in 1 h, followed by IL-2 and IL-12. There is no significant change for IL-6[57] and IL-10[29,57] in the liver. Especially, the level of IL-10[29] is very low in the liver compared with that in the plasma, which suggested that IL-10 might originate from other tissues, such as the spleen. But one previous paper reported that IL-10 expression in the liver is higher than that in the spleen[57]. As yet, where IL-10 originates remains unanswered.

Figure 2
Figure 2 Different cytokines levels within 24 h. A: Plasma level; B: Liver level. TNF: Tumor necrosis factor; IFN: Interferon; IL: Interleukin.

Comparing the acute and chronic animal models, the expression profiles of IL-10 are quite different. For example, in the acute model induced by Con A, TNF-α, IFN-γ and IL-12 levels increased to 2.11, 1.92 and 8.30 times of their normal level, respectively, after neutralization of IL-10. Reversely, administration of recombinant IL-10 prior to injection of Con A decreased by 47%, 47% and 80% of TNF-α, IFN-γ and IL-12 expression levels respectively. IL-10 is considered to be an anti-inflammatory cytokine in a murine model of Con A[58]. Kato et al[59] described that the IL-10 level is increased at 12 h after the Con A injection. After neutralizing antibodies to IL-10, it was intraperitoneally injected into animals of the same model at 6 h before Con A treatment, with serum alanine aminotransferase level being significantly higher than in the control group. Histological studies showed spotty necrosis in the group treated with anti-IL-10 antibodies. These results suggest that IL-10 has an inhibitory effect on liver injury in a murine model of Con A-induced experimental liver injury mediated by cellular immunity[58]. These studies suggested that both endogenous and exogenous IL-10 can protect the liver from acute injury[59].

However, there is evidence indicating that IL-10 could accelerate liver injury in the chronic model[60]. When Con A was administrated intravenously to BALB/c mice once a week, the IL-10 expression level in plasma increased to 7 times higher 20 wk later. Accordingly, in this model, inflammatory infiltration also lasted for 20 wk and activated stellate cells also dramatically increased[60]. All these results suggested that IL-10 aggravated liver injury in the chronic Con A model.

Paradoxically, IL-10 does not play the same role in all chronic models. For example, in the CCl4 chronic model, IL-10 slows down the process of fibrosis[61]. This may be due to the fact that the mechanisms of liver injury in these two models are different, and the latter does not involve T cell activation. In the acute Con A model, IL-10 may inhibit macrophages and Th1 cells from releasing inflammatory cytokines, which explains why it plays an anti-inflammation role in the acute model[58]. Though IL-10 can inhibit the secretion of anti-inflammation cytokines, secretion of IFN-γ is also inhibited[62]. Some previous studies reported that, to some extent, IFN-γ may relieve liver fibrosis. Therefore, a long duration of IFN-γ deficiency may aggravate fibrosis. As for the CCl4 model, liver injury is mediated only by free radicals, which is not relevant to the activation of the immune response and the release of inflammation cytokines. In conclusion, the expression profiles in different models, even with the same inducer, are not the same. The various mechanisms, cell types and micro-environments should be taken into consideration in experimental design and execution.

CON A MODEL AND NEW DRUG DEVELOPMENT

In recent years, based on the Con A animal model, many new therapeutic antibodies or proteins have been developed to attenuate liver injury in experimental models (Table 2)[63-66].

Table 2 New drugs developed based on the Concanavalin A model.
ClassificationTargetPathway
Hu 23C3[63]Monoclonal antibodyHuman osteopontinNF-κB
Anti-his H1[64]Polyclonal antibodyHistone H1NF-κB
ApoA II[65]High density lipoproteinLeukocytes and T cells-
CpG ODN[66]OligodeoxynucleotidesDNA binding ability of NF-κBNF-κB

Fan et al[63] humanized a murine monoclonal antibody 23C3 against human osteopontin by a complementary-determining region grafting method based on computer-assisted molecular modeling, denoted as Hu23C3. They demonstrated that Hu23C3 could have the potential for attenuating Con A-induced liver injury through the nuclear factor kappa B (NF-κB) pathway.

Nakano et al[64] intraperitoneally injected a polyclonal antibody against histone H1 immediately after Con A injection; they found that injection of anti-histone H1 antibodies could reduce Con A-induced liver damage, also via the NF-κB pathway.

It is reported that Con A-induced hepatitis was attenuated by the administration of apolipoprotein A-II, which is the second major apolipoprotein of high-density lipoprotein[65]; this inhibited leukocytes infiltration and the expression of T-cell related cytokines and chemokines.

The survival rate of mice was markedly enhanced by the administration of CpG-containing oligodeoxynucleotides (CpG ODN)[66]. This is because CpG ODN pretreatment inhibits the DNA binding ability of NF-κB, leading to the decrease of systemic/liver levels of TNF-α and IFN-γ. These results suggest that CpG ODN pretreatment protects the mice from Con A-induced liver injury, also via NF-κB pathway.

CONCLUSION

In this article we reviewed the evolution of the AIH model and emphasized the importance of the Con A AIH model. Based on the previous papers, we summarized the mechanisms of Con A-induced liver injury, its pathogenic changes and cytokines expression levels. The Con A animal model, which is a typical T cell dependent model, can mimic the mechanisms of clinical AIH diseases. Therefore, we think that it is a good and convenient model for studying the mechanisms of AIH and developing new therapeutic drugs.

Footnotes

Peer reviewer: Yoshiaki Iwasaki, MD, PhD, Associate Pro-fessor, Health Service Center, Okayama University, 2-1-1, Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan

S- Editor Sun H L- Editor Rutherford A E- Editor Li JY

References
1.  Manns MP, Vogel A. Autoimmune hepatitis, from mechanisms to therapy. Hepatology. 2006;43:S132-S144.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 133]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
2.  Primo J, Maroto N, Martínez M, Antón MD, Zaragoza A, Giner R, Devesa F, Merino C, del Olmo JA. Incidence of adult form of autoimmune hepatitis in Valencia (Spain). Acta Gastroenterol Belg. 2009;72:402-406.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Clark JM, Brancati FL, Diehl AM. The prevalence and etiology of elevated aminotransferase levels in the United States. Am J Gastroenterol. 2003;98:960-967.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Czaja AJ, Carpenter HA. Distinctive clinical phenotype and treatment outcome of type 1 autoimmune hepatitis in the elderly. Hepatology. 2006;43:532-538.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 152]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
5.  Al-Chalabi T, Underhill JA, Portmann BC, McFarlane IG, Heneghan MA. Impact of gender on the long-term outcome and survival of patients with autoimmune hepatitis. J Hepatol. 2008;48:140-147.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Häyry P, Gannedahl G. [Transplantation. Immunosuppression]. Nord Med. 1994;109:191-193.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Yang L, Liu YF, Liu SR, Wu G, Zhang JL, Meng YM, Shong SW, Li GC. Prevention and treatment of rejection after simultaneous pancreas-kidney transplantation. Chin Med Sci J. 2005;20:210-213.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Heneghan MA, McFarlane IG. Current and novel immunosuppressive therapy for autoimmune hepatitis. Hepatology. 2002;35:7-13.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Czaja AJ, Menon KV, Carpenter HA. Sustained remission after corticosteroid therapy for type 1 autoimmune hepatitis: a retrospective analysis. Hepatology. 2002;35:890-897.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  van Mourik ID, Kelly DA. Immunosuppressive drugs in paediatric liver transplantation. Paediatr Drugs. 2001;3:43-60.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Malekzadeh R, Nasseri-Moghaddam S, Kaviani MJ, Taheri H, Kamalian N, Sotoudeh M. Cyclosporin A is a promising alternative to corticosteroids in autoimmune hepatitis. Dig Dis Sci. 2001;46:1321-1327.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Czaja AJ. Rapidity of treatment response and outcome in type 1 autoimmune hepatitis. J Hepatol. 2009;51:161-167.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Teufel A, Weinmann A, Kahaly GJ, Centner C, Piendl A, Wörns M, Lohse AW, Galle PR, Kanzler S. Concurrent autoimmune diseases in patients with autoimmune hepatitis. J Clin Gastroenterol. 2010;44:208-213.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 137]  [Cited by in F6Publishing: 124]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
14.  Danielsson A, Prytz H. Oral budesonide for treatment of autoimmune chronic active hepatitis. Aliment Pharmacol Ther. 1994;8:585-590.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Czaja AJ, Freese DK. Diagnosis and treatment of autoimmune hepatitis. Hepatology. 2002;36:479-497.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Czaja AJ. Animal models of autoimmune hepatitis. Expert Rev Gastroenterol Hepatol. 2010;4:429-443.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 22]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
17.  Buschenfelde KH, Kossling FK, Miescher PA. Experimental chronic active hepatitis in rabbits following immunization with human liver proteins. Clin Exp Immunol. 1972;11:99-108.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Mihas AA, Subramony C, Achord JL. Experimental autoimmune hepatitis in mice following immunization with syngeneic liver proteins. J Med. 1995;26:309-322.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Lohse AW, Manns M, Dienes HP, Meyer zum Büschenfelde KH, Cohen IR. Experimental autoimmune hepatitis: disease induction, time course and T-cell reactivity. Hepatology. 1990;11:24-30.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Tiegs G, Hentschel J, Wendel A. A T cell-dependent experimental liver injury in mice inducible by concanavalin A. J Clin Invest. 1992;90:196-203.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Kim WH, Hong F, Radaeva S, Jaruga B, Fan S, Gao B. STAT1 plays an essential role in LPS/D-galactosamine-induced liver apoptosis and injury. Am J Physiol Gastrointest Liver Physiol. 2003;285:G761-G768.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
22.  Jirillo E, Caccavo D, Magrone T, Piccigallo E, Amati L, Lembo A, Kalis C, Gumenscheimer M. The role of the liver in the response to LPS: experimental and clinical findings. J Endotoxin Res. 2002;8:319-327.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 67]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
23.  El Hefnawi MM, El Behaidy WH, Youssif AA, Ghalwash AZ, El Housseiny LA, Zada S. Natural genetic engineering of hepatitis C virus NS5A for immune system counterattack. Ann N Y Acad Sci. 2009;1178:173-185.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Pearson T, Greiner DL, Shultz LD. Creation of "humanized" mice to study human immunity. Curr Protoc Immunol. 2008;Chapter 15:Unit 15.21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 135]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
25.  Guan Q, Ma Y, Hillman CL, Ma A, Zhou G, Qing G, Peng Z. Development of recombinant vaccines against IL-12/IL-23 p40 and in vivo evaluation of their effects in the downregulation of intestinal inflammation in murine colitis. Vaccine. 2009;27:7096-7104.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Autoantigen-specific regulatory T cells, a potential tool for immune-tolerance reconstitution in type-2 autoimmune hepatitis Hepatology. 2010;53:536-547.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 86]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
27.  Christen U, Holdener M, Hintermann E. Cytochrome P450 2D6 as a model antigen. Dig Dis. 2010;28:80-85.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Lapierre P, Djilali-Saiah I, Vitozzi S, Alvarez F. A murine model of type 2 autoimmune hepatitis: Xenoimmunization with human antigens. Hepatology. 2004;39:1066-1074.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 124]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
29.  Sass G, Heinlein S, Agli A, Bang R, Schümann J, Tiegs G. Cytokine expression in three mouse models of experimental hepatitis. Cytokine. 2002;19:115-120.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Tsui TY, Obed A, Siu YT, Yet SF, Prantl L, Schlitt HJ, Fan ST. Carbon monoxide inhalation rescues mice from fulminant hepatitis through improving hepatic energy metabolism. Shock. 2007;27:165-171.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
31.  Bertoletti A, Maini MK. Protection or damage: a dual role for the virus-specific cytotoxic T lymphocyte response in hepatitis B and C infection? Curr Opin Microbiol. 2000;3:387-392.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Hunter AL, Holscher MA, Neal RA. Thioacetamide-induced hepatic necrosis. I. Involvement of the mixed-function oxidase enzyme system. J Pharmacol Exp Ther. 1977;200:439-448.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Low TY, Leow CK, Salto-Tellez M, Chung MC. A proteomic analysis of thioacetamide-induced hepatotoxicity and cirrhosis in rat livers. Proteomics. 2004;4:3960-3974.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 113]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
34.  Thiele GM, Duryee MJ, Willis MS, Tuma DJ, Radio SJ, Hunter CD, Schaffert CS, Klassen LW. Autoimmune hepatitis induced by syngeneic liver cytosolic proteins biotransformed by alcohol metabolites. Alcohol Clin Exp Res. 2010;34:2126-2136.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 40]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
35.  Soares PA, Nascimento CO, Porto TS, Correia MT, Porto AL, Carneiro-da-Cunha MG. Purification of a lectin from Canavalia ensiformis using PEG-citrate aqueous two-phase system. J Chromatogr B Analyt Technol Biomed Life Sci. 2011;879:457-460.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Kanellopoulos PN, Pavlou K, Perrakis A, Agianian B, Vorgias CE, Mavrommatis C, Soufi M, Tucker PA, Hamodrakas SJ. The crystal structure of the complexes of concanavalin A with 4'-nitrophenyl-alpha-D-mannopyranoside and 4'-nitrophenyl-alpha-D-glucopyranoside. J Struct Biol. 1996;116:345-355.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Sato K, Imoto Y, Sugama J, Seki S, Inoue H, Odagiri T, Anzai J. Sugar-sensitive thin films composed of concanavalin A and glycogen. Anal Sci. 2004;20:1247-1248.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Fayad R, Sennello JA, Kim SH, Pini M, Dinarello CA, Fantuzzi G. Induction of thymocyte apoptosis by systemic administration of concanavalin A in mice: role of TNF-alpha, IFN-gamma and glucocorticoids. Eur J Immunol. 2005;35:2304-2312.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 29]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
39.  Carambia A, Herkel J. CD4 T cells in hepatic immune tolerance. J Autoimmun. 2010;34:23-28.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Zhang X, Wei HX, Rui S, Wei H, Tian Z. Opposite effects of high and low doses of interleukin-2 on T cell-mediated hepatitis in mice (interleukin-2 on hepatitis). Hepatol Int. 2010;4:641-648.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
41.  Varthaman A, Khallou-Laschet J, Clement M, Fornasa G, Kim HJ, Gaston AT, Dussiot M, Caligiuri G, Herbelin A, Kaveri S. Control of T cell reactivation by regulatory Qa-1-restricted CD8 T cells. J Immunol. 2010;184:6585-6591.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Zheng ZY, Weng SY, Yu Y. Signal molecule-mediated hepatic cell communication during liver regeneration. World J Gastroenterol. 2009;15:5776-5783.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  van der Meer W, van Gelder W, de Keijzer R, Willems H. The divergent morphological classification of variant lymphocytes in blood smears. J Clin Pathol. 2007;60:838-839.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Shi X, Zhang P, Sempowski GD, Shellito JE. Thymopoietic and bone marrow response to murine Pneumocystis pneumonia. Infect Immun. 2011;79:2031-2042.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  de Winther MP, Heeringa P. Bone marrow transplantations to study gene function in hematopoietic cells. Methods Mol Biol. 2011;693:309-320.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
46.  Peaudecerf L, dos Santos PR, Boudil A, Ezine S, Pardigon N, Rocha B. The role of the gut as a primary lymphoid organ: CD8αα intraepithelial T lymphocytes in euthymic mice derive from very immature CD44+ thymocyte precursors. Mucosal Immunol. 2011;4:93-101.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Tadmor T, Zhang Y, Cho HM, Podack ER, Rosenblatt JD. The absence of B lymphocytes reduces the number and function of T-regulatory cells and enhances the anti-tumor response in a murine tumor model. Cancer Immunol Immunother. 2011;60:609-619.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 96]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
48.  Repellin CE, Tsimbouri PM, Philbey AW, Wilson JB. Lymphoid hyperplasia and lymphoma in transgenic mice expressing the small non-coding RNA, EBER1 of Epstein-Barr virus. PLoS One. 2010;5:e9092.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
49.  Final Report on Carcinogens Background Document for Formaldehyde Rep Carcinog Backgr Doc. 2010;i-512.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Kage M. Pathology of autoimmune liver diseases in children. Hepatol Res. 2007;37 Suppl 3:S502-S508.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Schümann J, Wolf D, Pahl A, Brune K, Papadopoulos T, van Rooijen N, Tiegs G. Importance of Kupffer cells for T-cell-dependent liver injury in mice. Am J Pathol. 2000;157:1671-1683.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Knolle PA, Gerken G, Loser E, Dienes HP, Gantner F, Tiegs G, Meyer zum Buschenfelde KH, Lohse AW. Role of sinusoidal endothelial cells of the liver in concanavalin A-induced hepatic injury in mice. Hepatology. 1996;24:824-829.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Wang J, Leclercq I, Brymora JM, Xu N, Ramezani-Moghadam M, London RM, Brigstock D, George J. Kupffer cells mediate leptin-induced liver fibrosis. Gastroenterology. 2009;137:713-723.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Gove ME, Rhodes DH, Pini M, van Baal JW, Sennello JA, Fayad R, Cabay RJ, Myers MG, Fantuzzi G. Role of leptin receptor-induced STAT3 signaling in modulation of intestinal and hepatic inflammation in mice. J Leukoc Biol. 2009;85:491-496.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Takahashi K, Murakami M, Kikuchi H, Oshima Y, Kubohara Y. Derivatives of Dictyostelium differentiation-inducing factors promote mitogen-activated IL-2 production via AP-1 in Jurkat cells. Life Sci. 2011;88:480-485.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Miller ML, Sun Y, Fu YX. Cutting edge: B and T lymphocyte attenuator signaling on NKT cells inhibits cytokine release and tissue injury in early immune responses. J Immunol. 2009;183:32-36.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Küsters S, Gantner F, Künstle G, Tiegs G. Interferon gamma plays a critical role in T cell-dependent liver injury in mice initiated by concanavalin A. Gastroenterology. 1996;111:462-471.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Di Marco R, Xiang M, Zaccone P, Leonardi C, Franco S, Meroni P, Nicoletti F. Concanavalin A-induced hepatitis in mice is prevented by interleukin (IL)-10 and exacerbated by endogenous IL-10 deficiency. Autoimmunity. 1999;31:75-83.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Kato M, Ikeda N, Matsushita E, Kaneko S, Kobayashi K. Involvement of IL-10, an anti-inflammatory cytokine in murine liver injury induced by Concanavalin A. Hepatol Res. 2001;20:232-243.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Louis H, Le Moine A, Quertinmont E, Peny MO, Geerts A, Goldman M, Le Moine O, Devière J. Repeated concanavalin A challenge in mice induces an interleukin 10-producing phenotype and liver fibrosis. Hepatology. 2000;31:381-390.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Louis H, Van Laethem JL, Wu W, Quertinmont E, Degraef C, Van den Berg K, Demols A, Goldman M, Le Moine O, Geerts A. Interleukin-10 controls neutrophilic infiltration, hepatocyte proliferation, and liver fibrosis induced by carbon tetrachloride in mice. Hepatology. 1998;28:1607-1615.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Tsukamoto H. Is interleukin-10 antifibrogenic in chronic liver injury? Hepatology. 1998;28:1707-1709.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Fan K, Zhang B, Yang H, Wang H, Tan M, Hou S, Qian W, Li B, Wang H, Dai J. A humanized anti-osteopontin antibody protects from Concanavalin A induced-liver injury in mice. Eur J Pharmacol. 2011;657:144-151.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Nakano T, Goto S, Lai CY, Hsu LW, Takaoka Y, Kawamoto S, Chiang KC, Shimada Y, Ohmori N, Goto T. Immunological aspects and therapeutic significance of an autoantibody against histone H1 in a rat model of concanavalin A-induced hepatitis. Immunology. 2010;129:547-555.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Yamashita J, Iwamura C, Sasaki T, Mitsumori K, Ohshima K, Hada K, Hara N, Takahashi M, Kaneshiro Y, Tanaka H. Apolipoprotein A-II suppressed concanavalin A-induced hepatitis via the inhibition of CD4 T cell function. J Immunol. 2011;186:3410-3420.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Zhang H, Gong Q, Li JH, Kong XL, Tian L, Duan LH, Tong J, Song FF, Fang M, Zheng F. CpG ODN pretreatment attenuates concanavalin A-induced hepatitis in mice. Int Immunopharmacol. 2010;10:79-85.  [PubMed]  [DOI]  [Cited in This Article: ]