Review Open Access
Copyright ©2012 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastroenterol. Dec 28, 2012; 18(48): 7141-7148
Published online Dec 28, 2012. doi: 10.3748/wjg.v18.i48.7141
Role of autoimmunity in primary biliary cirrhosis
Tian-Yan Shi, Feng-Chun Zhang, Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing 100032, China
Author contributions: Shi TY and Zhang FC designed the research; Shi TY wrote the manuscript; and Zhang FC edited the manuscript.
Correspondence to: Feng-Chun Zhang, MD, Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, No. 41 Da Mu Cang, Western District, Beijing 100032, China. zhangfccra@hotmail.com
Telephone: +86-10-88068795 Fax: +86-10-88068794
Received: April 28, 2012
Revised: July 16, 2012
Accepted: August 14, 2012
Published online: December 28, 2012

Abstract

Primary biliary cirrhosis (PBC) is an autoimmune liver disease characterized by the presence of serum autoantibodies and chronic nonsuppurative destructive cholangitis. The pathogenesis of PBC involves environmental factors, genetic predisposition and loss of immune tolerance. In recent years, it has become univocally accepted that an inappropriately activated immune response is one of the most important factors in PBC. In this study, the role of autoimmunity in PBC is summarized and a feasible research orientation is recommended.

Key Words: Primary biliary cirrhosis, Autoimmunity, Humoral immunity, Cellular immunity, Nonspecific immunity



INTRODUCTION

Primary biliary cirrhosis (PBC) is an organ-specific autoimmune disease of the liver characterized by the presence of serum antimitochondrial antibodies (AMAs) and the destruction of small and medium-sized bile ducts. The histological manifestations are damaged biliary epithelial cells (BECs) and infiltration of T cells, B cells, macrophages, eosinophils and natural killer (NK) cells in the portal area, which eventually leads to cirrhosis and liver failure[1]. PBC affects middle-aged women, and the natural disease history is 10 to 20 year. The annual incidence rates range between 0.7 and 49 cases per million persons, while the prevalence rates range between 6.7 and 402 cases per million persons[2,3]. Advanced biochemical assays and improved acquisition of disease will lead to increased detection worldwide. Currently, the only recommended first-line therapy, early treatment with ursodeoxycholic acid (UDCA) at a dose of 13-15 mg/kg per day, can delay progression of histology and ameliorate long-term prognosis[4]. However, approximately 25% of patients have no response to UDCA, and in these cases liver transplantation is needed[5]. Exploration of pathogenesis is needed to discover novel target treatments. Deficiencies in autoimmune tolerance are critical factors of disease initiation and perpetuation. Therefore, the aim of this study was to demonstrate the function of autoimmunity in PBC.

HUMORAL IMMUNITY
Autoantibody-AMA

High titer of AMAs is the serological hallmark of PBC. It can be detected even before clinical symptoms or biochemical anomalies. The proportion of AMA-negative patients has been minimized due to the development of sensitive detection technology[6,7]. The highly disease-specific autoantibody, AMA-M2, recognizes components of the oxo-acid dehydrogenase complex (OADC) that are ubiquitously expressed on the inner mitochondrial membrane, including the E2 subset of the pyruvate dehydrogenase complex (PDC-E2). The antigens are released from apoptotic blebs of the BEC, or come from molecular mimicry of infectious agents, or from alteration of xenobiotics[8]. However, transgenic mice aberrantly expressing PDC-E2 components on BECs do not show serological and histological features of PBC[9], indicating that aberrant PDC-E2 expression is not sufficient for disease development.

Although AMA is found in most PBC patients, it may have no pathogenic role due to the following observations: (1) the titer of AMA is not associated with biochemical or histological manifestations, and there is no indication for a significant change in AMA levels after drug therapy[10,11]; (2) approximately 5% of patients are AMA-negative with typical histological injury and exhibit the same treatment response as AMA-positive patients[12,13]; and (3) although the antigens are ubiquitous, the disease is organ-specific.

However, some studies have identified novel features of AMAs in recent years. Lleo and colleagues[14,15] demonstrated that PDC-E2 with antigenic reactivity was only detectable in apoptotic blebs of human intra-hepatic BECs; Moreover, in the presence of AMA, macrophages increased the expression of TNF-related apoptosis-inducing ligand (TRAIL) and produced intense inflammatory cytokines, such as tumor necrosis factor alpha (TNF-α) and interleukin (IL)-6. These results provide a mechanism for the biliary specificity and a physiopathologic role of AMA in PBC. Rigopoulou et al[16] found that AMA-IgG3 was associated with a more severe disease. The presence of IgA-anti-PDC-E2 in sera or saliva might be associated with the progression of PBC[17]. The mechanism responsible is likely that a greater concentration of IgA in bile ducts can make cells more susceptible to apoptosis through constant transcytosis, resulting in subsequent bile duct damage[18]. In addition, the production of AMA-IgM from peripheral blood mononuclear cells (PBMCs) from PBC patients was reduced after exposure to UDCA[19].

Autoantibody-antinuclear antibodies

In addition to AMA, serum antinuclear antibodies (ANAs) are positive in approximately 1/3 patients with PBC. These antibodies provide important evidence for AMA-negative PBC. The typical staining pattern and clinical significance[20-28] of ANAs are shown in Table 1.

Table 1 The characteristics of antinuclear antibodies in primary biliary cirrhosis patients.
Staining patternAutoantigenPrevalence (%)Clinical significance
Nuclear dotSP100[20,28]25Highly specific for PBC; Urinary tract infections
PML[21,28]19Highly specific for PBC; Coexistence with anti-sp100
SP140[22]15Coexistence with anti-sp100 and anti-PML
Nuclear peripherygp210[23]25Association with disease severity and poor prognosis
(Nuclear pore complex)p62[24]30-55Association with disease severity and poor prognosis
Nuclear peripheryLamin[25]6-8Not highly specific for PBC
(Nuclear envelope)LBR[24]2-6Highly specific for PBC
AnticentromereCentromere[26]30Association with portal hypertension
?AchR M3[27]83Unknown
B cells and plasma cells

Plasma cells that originate from B cells are sources of antibodies. As antigen presentation cells, B cells can secrete many kinds of cytokines and present costimulatory signals to active antigen-specific T lymphocytes. Migita et al[29] found that serum B-cell-activating factor (BAFF) levels were significantly higher in PBC patients than in healthy controls and HCV-infected patients, and were positively correlated with aspartate amino-transferase (AST) and total bilirubin levels. In liver, CD5+ and CD20+ cells were associated with BEC damage, suggesting that B cells have a role in regulating the portal destruction in PBC[30]. Therefore, B cell depletion therapy might be an alternative to UDCA. In murine experiments, Igμ(-/-)NOD.c3c4 mice demonstrated a decreased number of activated NK cells in the liver. The degree of granuloma formation, bile duct destruction, and salivary gland histology were also shown to be significantly attenuated[31]. Moreover, anti-CD20 therapy every 2 wk in transforming growth factor-beta receptor II dominant negative (dnTGF-βRII) mice at age of 4-6 wk could reduce the number of B cells and CD8+ T cells in liver[32]. In clinical therapy, two doses of 1000 mg rituximab separated by 2 wk were safe and effective in patients with an incomplete UDCA response. After treatment, not only did serum levels of total IgG, IgM, and IgA decrease significantly, but T regulatory (Treg) cells also increased, which was associated with increased mRNA levels of forkhead box 3 (Foxp3) and TGF-β in CD4+ T cells[33].

In contrast, in 2-octynoic acid-bovine serum albumin (2OA-BSA)-induced mice, treatment with anti-CD20 and anti-CD79 antibodies increased the number of CD4+ and CD8+ T cells infiltrating around damaged bile ducts in portal areas, leading to more severe cholangitis[34]. A similar phenomenon occurred in Igμ(-/-)dnTGF-βRII mice. Adoptive transfer of CD19+ cells from dnTGF-βRII mice into recombination activating gene-1 (Rag-1)(-/-) mice resulted in decreased liver inflammation and bile duct damage[35]. However, anti-CD20 therapy in dnTGF-βRII mice at age of 20-22 wk had little effect on liver lesions[32]. The efficacies of different B cell depletion approaches in diverse murine models are shown in Table 2. Together, these findings suggest that there is a subclass of B cells that have a regulatory role by producing IL-10[36]. However, further exploration of the function of B cells in PBC is needed before B cell depletion therapy can be applied to routine clinical work.

Table 2 Differences in B cell depletion therapies in primary biliary cirrhosis murine models.
ResultsModelTherapy (time)
AmeliorationNOD.c3c4 miceIgμ knockout
dnTGF-βRII miceAnti-CD20 (4-6 wk)
Exacerbation2OA-BSA-induced miceAnti-CD20/Anti-CD79 (6 wk)
dnTGF-βRII miceIgμ knockout
No effectdnTGF-βRII miceAnti-CD20 (20-22 wk)
Elevated IgM

In addition to high titers of circulating AMAs, PBC patients have high levels of serum IgM that are not related to titers of AMAs. Compared with AMA-positive PBC, the level of serum IgM was lower in AMA-negative patients[37,38]. Plasma cells in the portal tracts of PBC patients are found to be predominantly IgM-positive, while those cells predominantly express IgG in other forms of liver disease, such as autoimmune hepatitis and chronic hepatitis C[39,40]. It has been shown that after treatment with UDCA, the level of IgM decreases at both short-term and long-term follow-up[41], which is possibly due to a reduction in naïve B cell and IgM-memory B cell activation through down-regulation of the NF-κB signaling pathway[19]. The mechanism of IgM elevation is still unclear in PBC, but abnormal Ig class switching may be involved. Lleo et al[42] showed that IgM was inversely associated with levels of CD40L promoter methylation in CD4+ T cells, suggesting that the CD40-CD40L interaction is involved in the production of high amounts of IgM. Moreover, CD40L is an essential molecule involved in Ig class switching.

CELLULAR IMMUNITY

It is thought that activated CD4+ T cells can recognize peptide PDC-E2163-176, while activated CD8+ T cells can recognize peptide PDC-E2159-167 and PDC-E2165-174 in PBC. Moreover, a large number of autoreactive T lymphocytes infiltrate the portal area.

CD4+ T cells

Effector CD4+ T cells: In patients with PBC, it is well accepted that an enhanced ratio of Th1 to Th2 cells is one of the most important factors[43,44] in the onset of disease. After treatment with UDCA, serum interferon-gamma (IFN-γ)[45] and liver IL-2[45,46], which are Th1 cell-related cytokines, are decreased. However, the level of serum IFN-γ rebounded after 6 mo of therapy[45]. Recently, besides Th1 cells, studies have shown that IL-17+ cells accumulate around the damaged bile ducts[47,48], and BECs produce Th17-inducible cytokines (IL-6, IL-1β and IL-23) when stimulated with pathogen-associated molecular patterns (PAMPs)[47]. In addition, the ratio of Th17 to Tregs is enhanced in PBMCs[49].

Several experiments using murine models have indicated a central role of CD4+ T cells in the pathogenesis of PBC. NOD.c3c4-SCID mice can develop autoimmune cholangitis after adoptive transfer of splenocytes or CD4+ T cells[50]. In IL-2Rα(-/-) mice, marked aggregation of IL-17+ cells within portal tracts compared to the periphery has been demonstrated. Interestingly, CD4+ T cells from the livers of normal C57BL/6J mice can secrete higher levels of IL-17 compared to those from spleens, indicating the role of the liver microenvironment in Th17 induction[48].

It is currently unknown whether Th1 or Th17 cells are more important in the pathogenesis of the disease. IL-12p40(-/-)dnTGF-βRII mice have a dramatic reduction in histological autoimmune cholangitis and a significant decrease in the levels of intra-hepatic proinflammatory cytokines[51], while worsening hepatic histology and elevated inflammatory cytokine production have been observed in IL-6(-/-)dnTGF-βRII mice[52]. These findings might suggest that IL-12-inducible Th1 cells are more important than IL-6-inducible Th17 cells. However, the definitive conclusions on these mechanisms will require further investigation.

Treg cells: CD4+CD25high regulatory T cells play a critical role in self-tolerance and the prevention of autoimmune disease. Patients with PBC display a relative reduction of circulating CD4+CD25high Tregs compared to controls[53,54]. The frequency of circulating Tregs can increase after 1 year of treatment with UDCA[13]. However, the number of Foxp3+ cells is higher in the liver in PBC[53], which is most likely due to the localization of CD8+ T cell blasts in the liver portal area[55].

In addition to CD4+ Tregs, Bernuzzi et al[56] found that the CD8+ Treg (CD8+CD28-) population has striking phenotypic alterations, including decreased CD39 and increased CD127. Although CD8+ Tregs were not significantly different quantitatively between patients and healthy subjects, the in vitro induction of CD8+ Tregs by incubation with IL-10 was significantly reduced in PBC patients.

Murine models constructed by altering the signaling pathway of Treg cells can imitate PBC-like manifestations, which underscores the vital function of Tregs in disease. Zhang et al[57] demonstrated that Scurfy mice, which have complete ablation of Foxp3+ Tregs, exhibited a high titer of AMAs and elevated serum cytokines (TNF-α, IFN-γ, IL-6, IL-12p40, and IL-23). In addition, these mice also had a substantial number of lymphocytes infiltrated around portal areas with bile duct damage. Both IL-2Rα(-/-)[58] and dnTGF-βRII murine models[59] show PBC-like characteristics.

CD8+ T cells

Compared to CD4+ T cells, CD8+ T cells play a more significant role in mediating the destruction of the bile duct. In PBC patients, CD45ROhighCD57+CD8high T cells expressing elevated α4β7 and IL-5 accumulate around the portal area[60]. Moreover, the ratio of CD8+ T cells to FoxP3+ cells is significantly higher in the liver at late stages of the disease[53]. Biliary ductule damage by CD8+ T cells is also observed in non-obese diabetic autoimmune biliary disease (NOD.ABD) mice[61]. CD8+ T cells from NOD.ABD mice can transfer liver inflammation into NOD.c3c4-SCID recipients. The liver of dnTGF-βRII mice has an increased number of CD8+ T cells and a higher ratio of CD8 to CD3 or CD4 cells[59]. Furthermore, adoptive transfer of CD8+ T cells from dnTGF-βRII mice to Rag-1(-/-) recipients results in PBC-like liver lesions and AMA-positive sera, while CD4+ T cells predominantly cause bowel inflammation[62]. Relative to IL-2Rα(-/-) mice, IL-2Rα(-/-) CD4(-/-) mice have increased biliary ductular damage. These findings also suggest that CD8+ T cells may mediate bile ductular injury under the background of Treg function loss[63].

INNATE IMMUNITY

The innate immune system is the first line of defense against infection. Emerging evidence suggests that the liver is one component of the body’s immune system[64]. Traditional immunosuppressive drugs are not effective for PBC compared to other autoimmune diseases. Thus, the role of the innate immune system in PBC has garnered a lot of focus in recent years.

Toll-like receptor

The liver can encounter a number of pathogenic microorganisms and their by-products from the gut by acting as a traffic hub. The expression of toll-like receptor (TLR) is normally regulated by a negative signaling pathway in the liver, which prevents inappropriate activation of inflammation.

After in vitro challenge with TLR ligands, circulating monocytes in PBC patients produce higher pro-inflammatory cytokines, including IL-1β, IL-6, IL-8, and TNF-α[65]. The expressions of TLR3[66] and TLR4[67,68] are increased in BECs. Transfection of poly(I:C) into BECs induces a marked increase in mRNAs encoding a variety of chemokines/cytokines[66]. In addition, the expressions of TLR4 in BECs[67] and circulating monocytes[69] both increase significantly after stimulation with lipopolysaccharide (LPS),. Furthermore, the level of RP105, which is involved in the negative regulation of TLR4 signaling, is decreased in PBC monocytes[69]. The interplay between LPS and TLR4 results in myeloid differentiation factor 88 (MyD88) recruitment, NF-κB activation and coding gene expression[67,69]. After exposure to CpG, PBMCs from PBC patients produce higher levels of IgM and AMAs[70,71]. Importantly, the effect can be inhibited by K+ channel blockers[70] and UDCA[19].

These results indicate that patients with PBC exhibit hypersensitivity of TLR signaling, which leads to a breakdown of self-tolerance. However, activation of the TLR signal pathway alone may not explain the entire course of the disease[72]. Mice immunized with 2OA-BSA coupled with poly (I:C) showed alterations in the disease process and acceleration of fibrosis of liver.

NKT cells

NKT cells are a subset of lymphocytes possessing both T cell receptors (TCRs) and NK-specific receptors, and they play an important part in the modulation of the innate immune response and cytotoxicity.

Alpha-galactosylceramide (α-GalCer) is an activator of NKT cells, and the frequency of CD1d-α-GalCer-restricted NKT cells is similar between the peripheral blood and liver of healthy people. In contrast, the frequency of these cells in liver is significantly higher than in peripheral blood in PBC patients, and also higher than in healthy individuals[73].

To define the function of CD1d-restricted NKT cells in the pathogenesis of PBC, Chuang et al[74] generated CD1d(-/-)dnTGF-βRII mice and found that they developed decreased hepatic lymphoid cell infiltration and mild cholangitis. After immunization of 2-OA-BSA-induced PBC mice with α-GalCer, Wu et al[75] found that the disease was exacerbated, including signs of portal inflammation, bile duct destruction and liver fibrosis. However, in vivo depletion of NK and NKT cells in the same murine model only suppressed AMAs and cytokine production, but did not change the portal cholangitis[76]. Therefore, these data support the role of NK and NKT cells in the loss of autoimmune tolerance in PBC; however, the development of PBC also requires other pathological factors.

NK cells

NK cells are another component of the innate immune system, and function by secreting cytokines and lysing target cells. NK cells account for 30% of the total resident lymphocytes in the liver. Chuang et al[77] reported an obvious higher frequency and absolute number of NK cells in both the blood and liver of PBC patients. Moreover, the cytotoxic activity and perforin expression of isolated NK cells were increased. Recently, Shimoda et al[78] demonstrated that TLR4 ligand-stimulated NK cells destroyed autologous BECs in the presence of IFN-α synthesized by TLR 3 ligand-stimulated monocytes. In addition, there was an increased number of CD56+ cells scattered around the destroyed small bile ducts.

CHEMOKINES

Chemokines, which are 8-12 kDa heparin-binding cytokines, direct lymphocyte trafficking and positioning in tissues and play roles in modulating immune responses and shaping the severity of disease[79]. In PBC, infiltrating memory T cells are culprits regarding the destruction of bile ducts.

Chemokines, interferon-gamma-inducible protein-10 (IP-10) and monokine-induced by gamma interferon (MIG), are increased in plasma and the portal area in PBC patients compared to controls. Moreover, the frequency of CXCR3+ cells is higher in both PBMCs and injured bile ducts. Intriguingly, daughters and sisters of PBC patients demonstrate increased plasma levels of IP-10 and MIG, but the frequency of circulating CXCR3+ cells is normal[80]. Knockout of CXCR3 in Poly(I:C)-induced mice displayed a delayed and milder progression of cellular inflammation[81].

Other chemokines may also be involved in recruiting inflammatory cells into the liver in PBC. BECs are a source of chemokine production that attracts monocytes into the liver[82,83]. Stimulated by poly(I:C), BECs produce augmented CX3CL1 in the presence of monocytes pretreated with LPS through CD40-CD154 contact[83]. In addition, BECs pretreated with LPS also secrete elevated CX3CL1[83,84].

CONCLUSION

Substantial amounts of data to date have illustrated that autoimmunity plays a critical role in the pathogenesis of PBC. The adaptive immune response, the innate immune system and their interplay participate in the development of disease. However, there are clearly limitations and unanswered questions that still remain: (1) although some murine models have been established, they cannot imitate PBC in humans completely (Table 3); (2) in vitro experiments based on human samples may not completely reflect the endosomatic problems; and (3) the data to date are mainly based on PBMCs, which lack reliability without assessment in the local liver microenvironment. In order to address these problems, better murine models are needed. In the future, researchers must identify new mechanisms through the analysis of both in vivo and in vitro approaches for the development of effective treatment strategies for PBC.

Table 3 The characteristics of primary biliary cirrhosis murine models and primary biliary cirrhosis patients.
NOD.c3c4[50]dnTGF-βR II[59]IL-2Rα(-/-)[58,85]Ae2a,b(-/-)[86]2OA-BSA induced[87]Human
Disease onset8-20 wk4 wk4 wk6 mo?4-12 wk40-60 yr
AMA56%100%100%40-80%100%95%
ANA80%-90%(-)80%(-)?30%
CytokinesTh1 ↑Th1 ↑Th1, Th17 ↑Th1 ↑Th1 ↑Th1, Th17 ↑
Treg cells
IgIgG, IgM ↑IgA ↑IgG, IgA ↑IgG, IgM ↑?IgM ↑
Liver inflammation
CD8+/CD4+T
Treg??
Fibrosis?Slight?SlightSlightEvident in late stage
ComplicationBile cyst andColitisColitis??Thyroid disorder
Extrahepatic damageAnemiaSjogren syndrome
Footnotes

Peer reviewers: Metin Basaranoglu, MD, PhD, Associate Professor, Gastroenterology Division, Ankara Hospital, Sihhiye, Tr-06000 Ankara, Turkey; Emanuel K Manesis, MD, Professor of Medicine, Athens University School of Medicine, Liver Unit, Euroclinic, 19 Mavromateon Street, 1034 Athens, Greece

S- Editor Shi ZF L- Editor Logan S E- Editor Zhang DN

References
1.  Kaplan MM, Gershwin ME. Primary biliary cirrhosis. N Engl J Med. 2005;353:1261-1273.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 965]  [Cited by in F6Publishing: 889]  [Article Influence: 46.8]  [Reference Citation Analysis (0)]
2.  Prince MI, James OF. The epidemiology of primary biliary cirrhosis. Clin Liver Dis. 2003;7:795-819.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 99]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
3.  Lazaridis KN, Talwalkar JA. Clinical epidemiology of primary biliary cirrhosis: incidence, prevalence, and impact of therapy. J Clin Gastroenterol. 2007;41:494-500.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 44]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
4.  Kuiper EM, Hansen BE, de Vries RA, den Ouden-Muller JW, van Ditzhuijsen TJ, Haagsma EB, Houben MH, Witteman BJ, van Erpecum KJ, van Buuren HR. Improved prognosis of patients with primary biliary cirrhosis that have a biochemical response to ursodeoxycholic acid. Gastroenterology. 2009;136:1281-1287.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 321]  [Cited by in F6Publishing: 315]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
5.  Corpechot C, Abenavoli L, Rabahi N, Chrétien Y, Andréani T, Johanet C, Chazouillères O, Poupon R. Biochemical response to ursodeoxycholic acid and long-term prognosis in primary biliary cirrhosis. Hepatology. 2008;48:871-877.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 451]  [Cited by in F6Publishing: 434]  [Article Influence: 27.1]  [Reference Citation Analysis (0)]
6.  Oertelt S, Rieger R, Selmi C, Invernizzi P, Ansari AA, Coppel RL, Podda M, Leung PS, Gershwin ME. A sensitive bead assay for antimitochondrial antibodies: Chipping away at AMA-negative primary biliary cirrhosis. Hepatology. 2007;45:659-665.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 125]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
7.  Bizzaro N, Covini G, Rosina F, Muratori P, Tonutti E, Villalta D, Pesente F, Alessio MG, Tampoia M, Antico A. Overcoming a "probable" diagnosis in antimitochondrial antibody negative primary biliary cirrhosis: study of 100 sera and review of the literature. Clin Rev Allergy Immunol. 2012;42:288-297.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 58]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
8.  Selmi C, De Santis M, Cavaciocchi F, Gershwin ME. Infectious agents and xenobiotics in the etiology of primary biliary cirrhosis. Dis Markers. 2010;29:287-299.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 17]  [Reference Citation Analysis (0)]
9.  Inamura K, Tsuji H, Nakamoto Y, Suzuki M, Kaneko S. Transgenic mice aberrantly expressing pyruvate dehydrogenase complex E2 component on biliary epithelial cells do not show primary biliary cirrhosis. Clin Exp Immunol. 2006;145:93-100.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 12]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
10.  Benson GD, Kikuchi K, Miyakawa H, Tanaka A, Watnik MR, Gershwin ME. Serial analysis of antimitochondrial antibody in patients with primary biliary cirrhosis. Clin Dev Immunol. 2004;11:129-133.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 42]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
11.  Zhang W, Moritoki Y, Tsuneyama K, Yang GX, Ilan Y, Lian ZX, Gershwin ME. Beta-glucosylceramide ameliorates liver inflammation in murine autoimmune cholangitis. Clin Exp Immunol. 2009;157:359-364.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 17]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
12.  Hirschfield GM, Heathcote EJ. Antimitochondrial antibody-negative primary biliary cirrhosis. Clin Liver Dis. 2008;12:323-331; viii-ix.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 27]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
13.  Liu B, Shi XH, Zhang FC, Zhang W, Gao LX. Antimitochondrial antibody-negative primary biliary cirrhosis: a subset of primary biliary cirrhosis. Liver Int. 2008;28:233-239.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 38]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
14.  Lleo A, Selmi C, Invernizzi P, Podda M, Coppel RL, Mackay IR, Gores GJ, Ansari AA, Van de Water J, Gershwin ME. Apotopes and the biliary specificity of primary biliary cirrhosis. Hepatology. 2009;49:871-879.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 162]  [Cited by in F6Publishing: 149]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
15.  Lleo A, Bowlus CL, Yang GX, Invernizzi P, Podda M, Van de Water J, Ansari AA, Coppel RL, Worman HJ, Gores GJ. Biliary apotopes and anti-mitochondrial antibodies activate innate immune responses in primary biliary cirrhosis. Hepatology. 2010;52:987-998.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 161]  [Cited by in F6Publishing: 170]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
16.  Rigopoulou EI, Davies ET, Bogdanos DP, Liaskos C, Mytilinaiou M, Koukoulis GK, Dalekos GN, Vergani D. Antimitochondrial antibodies of immunoglobulin G3 subclass are associated with a more severe disease course in primary biliary cirrhosis. Liver Int. 2007;27:1226-1231.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 54]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
17.  Tanaka A, Nezu S, Uegaki S, Mikami M, Okuyama S, Kawamura N, Aiso M, Gershwin ME, Takahashi S, Selmi C. The clinical significance of IgA antimitochondrial antibodies in sera and saliva in primary biliary cirrhosis. Ann N Y Acad Sci. 2007;1107:259-270.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 28]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
18.  Matsumura S, Van De Water J, Leung P, Odin JA, Yamamoto K, Gores GJ, Mostov K, Ansari AA, Coppel RL, Shiratori Y. Caspase induction by IgA antimitochondrial antibody: IgA-mediated biliary injury in primary biliary cirrhosis. Hepatology. 2004;39:1415-1422.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 76]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
19.  Kikuchi K, Hsu W, Hosoya N, Moritoki Y, Kajiyama Y, Kawai T, Takai A, Hayami E, Selmi C, Gershwin ME. Ursodeoxycholic acid reduces CpG-induced IgM production in patients with primary biliary cirrhosis. Hepatol Res. 2009;39:448-454.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 23]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
20.  Manuel Lucena J, Montes Cano M, Luis Caro J, Respaldiza N, Alvarez A, Sánchez-Román J, Núñez-Roldán A, Wichmann I. Comparison of two ELISA assays for anti-Sp100 determination. Ann N Y Acad Sci. 2007;1109:203-211.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 6]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
21.  Züchner D, Sternsdorf T, Szostecki C, Heathcote EJ, Cauch-Dudek K, Will H. Prevalence, kinetics, and therapeutic modulation of autoantibodies against Sp100 and promyelocytic leukemia protein in a large cohort of patients with primary biliary cirrhosis. Hepatology. 1997;26:1123-1130.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 27]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
22.  Granito A, Yang WH, Muratori L, Lim MJ, Nakajima A, Ferri S, Pappas G, Quarneti C, Bianchi FB, Bloch DB. PML nuclear body component Sp140 is a novel autoantigen in primary biliary cirrhosis. Am J Gastroenterol. 2010;105:125-131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 67]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
23.  Ishibashi H, Komori A, Shimoda S, Ambrosini YM, Gershwin ME, Nakamura M. Risk factors and prediction of long-term outcome in primary biliary cirrhosis. Intern Med. 2011;50:1-10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 26]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
24.  Miyachi K, Hankins RW, Matsushima H, Kikuchi F, Inomata T, Horigome T, Shibata M, Onozuka Y, Ueno Y, Hashimoto E. Profile and clinical significance of anti-nuclear envelope antibodies found in patients with primary biliary cirrhosis: a multicenter study. J Autoimmun. 2003;20:247-254.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 75]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
25.  Nesher G, Margalit R, Ashkenazi YJ. Anti-nuclear envelope antibodies: Clinical associations. Semin Arthritis Rheum. 2001;30:313-320.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 34]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
26.  Nakamura M, Kondo H, Mori T, Komori A, Matsuyama M, Ito M, Takii Y, Koyabu M, Yokoyama T, Migita K. Anti-gp210 and anti-centromere antibodies are different risk factors for the progression of primary biliary cirrhosis. Hepatology. 2007;45:118-127.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 284]  [Cited by in F6Publishing: 247]  [Article Influence: 14.5]  [Reference Citation Analysis (0)]
27.  Berg CP, Blume K, Lauber K, Gregor M, Berg PA, Wesselborg S, Stein GM. Autoantibodies to muscarinic acetylcholine receptors found in patients with primary biliary cirrhosis. BMC Gastroenterol. 2010;10:120.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
28.  Mytilinaiou MG, Meyer W, Scheper T, Rigopoulou EI, Probst C, Koutsoumpas AL, Abeles D, Burroughs AK, Komorowski L, Vergani D. Diagnostic and clinical utility of antibodies against the nuclear body promyelocytic leukaemia and Sp100 antigens in patients with primary biliary cirrhosis. Clin Chim Acta. 2012;413:1211-1216.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 40]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
29.  Migita K, Ilyassova B, Kovzel EF, Nersesov A, Abiru S, Maeda Y, Komori A, Ito M, Yano K, Yatsuhashi H. Serum BAFF and APRIL levels in patients with PBC. Clin Immunol. 2010;134:217-225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 31]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
30.  Jin Q, Moritoki Y, Lleo A, Tsuneyama K, Invernizzi P, Moritoki H, Kikuchi K, Lian ZX, Hirschfield GM, Ansari AA. Comparative analysis of portal cell infiltrates in antimitochondrial autoantibody-positive versus antimitochondrial autoantibody-negative primary biliary cirrhosis. Hepatology. 2012;55:1495-1506.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 32]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
31.  Moritoki Y, Tsuda M, Tsuneyama K, Zhang W, Yoshida K, Lian ZX, Yang GX, Ridgway WM, Wicker LS, Ansari AA. B cells promote hepatic inflammation, biliary cyst formation, and salivary gland inflammation in the NOD.c3c4 model of autoimmune cholangitis. Cell Immunol. 2011;268:16-23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
32.  Moritoki Y, Lian ZX, Lindor K, Tuscano J, Tsuneyama K, Zhang W, Ueno Y, Dunn R, Kehry M, Coppel RL. B-cell depletion with anti-CD20 ameliorates autoimmune cholangitis but exacerbates colitis in transforming growth factor-beta receptor II dominant negative mice. Hepatology. 2009;50:1893-1903.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 73]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
33.  Tsuda M, Moritoki Y, Lian ZX, Zhang W, Yoshida K, Wakabayashi K, Yang GX, Nakatani T, Vierling J, Lindor K. Biochemical and immunologic effects of rituximab in patients with primary biliary cirrhosis and an incomplete response to ursodeoxycholic acid. Hepatology. 2012;55:512-521.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 123]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
34.  Dhirapong A, Lleo A, Yang GX, Tsuneyama K, Dunn R, Kehry M, Packard TA, Cambier JC, Liu FT, Lindor K. B cell depletion therapy exacerbates murine primary biliary cirrhosis. Hepatology. 2011;53:527-535.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 61]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
35.  Moritoki Y, Zhang W, Tsuneyama K, Yoshida K, Wakabayashi K, Yang GX, Bowlus C, Ridgway WM, Ueno Y, Ansari AA. B cells suppress the inflammatory response in a mouse model of primary biliary cirrhosis. Gastroenterology. 2009;136:1037-1047.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 69]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
36.  Fillatreau S, Gray D, Anderton SM. Not always the bad guys: B cells as regulators of autoimmune pathology. Nat Rev Immunol. 2008;8:391-397.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 213]  [Cited by in F6Publishing: 212]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
37.  Zhang FK, Jia JD, Wang BE. Clinical evaluation of serum antimitochondrial antibody-negative primary biliary cirrhosis. Hepatobiliary Pancreat Dis Int. 2004;3:288-291.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Hu C, Deng C, Song G, Zhang W, Zhang S, Li X, Li P, Zhang F, Li Y. Prevalence of autoimmune liver disease related autoantibodies in Chinese patients with primary biliary cirrhosis. Dig Dis Sci. 2011;56:3357-3363.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 20]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
39.  Daniels JA, Torbenson M, Anders RA, Boitnott JK. Immunostaining of plasma cells in primary biliary cirrhosis. Am J Clin Pathol. 2009;131:243-249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 28]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
40.  Moreira RK, Revetta F, Koehler E, Washington MK. Diagnostic utility of IgG and IgM immunohistochemistry in autoimmune liver disease. World J Gastroenterol. 2010;16:453-457.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 33]  [Cited by in F6Publishing: 27]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
41.  Kuiper EM, Hansen BE, Lesterhuis W, Robijn RJ, Thijs JC, Engels LG, Koek GH, Aparicio MN, Kerbert-Dreteler MJ, van Buuren HR. The long-term effect of ursodeoxycholic acid on laboratory liver parameters in biochemically non-advanced primary biliary cirrhosis. Clin Res Hepatol Gastroenterol. 2011;35:29-33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 18]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
42.  Lleo A, Liao J, Invernizzi P, Zhao M, Bernuzzi F, Ma L, Lanzi G, Ansari AA, Coppel RL, Zhang P. Immunoglobulin M levels inversely correlate with CD40 ligand promoter methylation in patients with primary biliary cirrhosis. Hepatology. 2012;55:153-160.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 105]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
43.  Harada K, Nakanuma Y. Molecular mechanisms of cholangiopathy in primary biliary cirrhosis. Med Mol Morphol. 2006;39:55-61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 26]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
44.  Harada K, Tsuneyama K, Yasoshima M, Kanemori Y, Ohta H, Masuda S, Onai N, Matsushima K, Nakanuma Y. Type1 and type2 memory T cells imbalance shown by expression of intrahepatic chemokine receptors relates to pathogenesis of primary biliary cirrhosis. Hepatol Res. 2002;24:290.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 15]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
45.  Tang M, Shi XH, Zhang FC. [The characteristics of peripheral lymphocytic subsets and cytokines in primary biliary and their changes to drug treatment.]. Zhonghua Neike Zazhi. 2010;49:129-133.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Miyaguchi S, Mori M. Ursodeoxycholic acid (UDCA) suppresses liver interleukin 2 mRNA in the cholangitis model. Hepatogastroenterology. 2005;52:596-602.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Harada K, Shimoda S, Sato Y, Isse K, Ikeda H, Nakanuma Y. Periductal interleukin-17 production in association with biliary innate immunity contributes to the pathogenesis of cholangiopathy in primary biliary cirrhosis. Clin Exp Immunol. 2009;157:261-270.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 137]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
48.  Lan RY, Salunga TL, Tsuneyama K, Lian ZX, Yang GX, Hsu W, Moritoki Y, Ansari AA, Kemper C, Price J. Hepatic IL-17 responses in human and murine primary biliary cirrhosis. J Autoimmun. 2009;32:43-51.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 145]  [Cited by in F6Publishing: 140]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
49.  Rong G, Zhou Y, Xiong Y, Zhou L, Geng H, Jiang T, Zhu Y, Lu H, Zhang S, Wang P. Imbalance between T helper type 17 and T regulatory cells in patients with primary biliary cirrhosis: the serum cytokine profile and peripheral cell population. Clin Exp Immunol. 2009;156:217-225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 126]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
50.  Irie J, Wu Y, Wicker LS, Rainbow D, Nalesnik MA, Hirsch R, Peterson LB, Leung PS, Cheng C, Mackay IR. NOD.c3c4 congenic mice develop autoimmune biliary disease that serologically and pathogenetically models human primary biliary cirrhosis. J Exp Med. 2006;203:1209-1219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 152]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
51.  Yoshida K, Yang GX, Zhang W, Tsuda M, Tsuneyama K, Moritoki Y, Ansari AA, Okazaki K, Lian ZX, Coppel RL. Deletion of interleukin-12p40 suppresses autoimmune cholangitis in dominant negative transforming growth factor beta receptor type II mice. Hepatology. 2009;50:1494-1500.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 106]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
52.  Zhang W, Tsuda M, Yang GX, Tsuneyama K, Rong G, Ridgway WM, Ansari AA, Flavell RA, Coppel RL, Lian ZX. Deletion of interleukin-6 in mice with the dominant negative form of transforming growth factor beta receptor II improves colitis but exacerbates autoimmune cholangitis. Hepatology. 2010;52:215-222.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 27]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
53.  Lan RY, Cheng C, Lian ZX, Tsuneyama K, Yang GX, Moritoki Y, Chuang YH, Nakamura T, Saito S, Shimoda S. Liver-targeted and peripheral blood alterations of regulatory T cells in primary biliary cirrhosis. Hepatology. 2006;43:729-737.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 236]  [Cited by in F6Publishing: 220]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
54.  Wang D, Zhang H, Liang J, Gu Z, Zhou Q, Fan X, Hou Y, Sun L. CD4+ CD25+ but not CD4+ Foxp3+ T cells as a regulatory subset in primary biliary cirrhosis. Cell Mol Immunol. 2010;7:485-490.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
55.  Bochtler P, Riedl P, Gomez I, Schirmbeck R, Reimann J. Local accumulation and activation of regulatory Foxp3+ CD4 T(R) cells accompanies the appearance of activated CD8 T cells in the liver. Hepatology. 2008;48:1954-1963.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 11]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
56.  Bernuzzi F, Fenoglio D, Battaglia F, Fravega M, Gershwin ME, Indiveri F, Ansari AA, Podda M, Invernizzi P, Filaci G. Phenotypical and functional alterations of CD8 regulatory T cells in primary biliary cirrhosis. J Autoimmun. 2010;35:176-180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 58]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
57.  Zhang W, Sharma R, Ju ST, He XS, Tao Y, Tsuneyama K, Tian Z, Lian ZX, Fu SM, Gershwin ME. Deficiency in regulatory T cells results in development of antimitochondrial antibodies and autoimmune cholangitis. Hepatology. 2009;49:545-552.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 79]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
58.  Wakabayashi K, Lian ZX, Moritoki Y, Lan RY, Tsuneyama K, Chuang YH, Yang GX, Ridgway W, Ueno Y, Ansari AA. IL-2 receptor alpha(-/-) mice and the development of primary biliary cirrhosis. Hepatology. 2006;44:1240-1249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 183]  [Cited by in F6Publishing: 193]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
59.  Oertelt S, Lian ZX, Cheng CM, Chuang YH, Padgett KA, He XS, Ridgway WM, Ansari AA, Coppel RL, Li MO. Anti-mitochondrial antibodies and primary biliary cirrhosis in TGF-beta receptor II dominant-negative mice. J Immunol. 2006;177:1655-1660.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Tsuda M, Ambrosini YM, Zhang W, Yang GX, Ando Y, Rong G, Tsuneyama K, Sumida K, Shimoda S, Bowlus CL. Fine phenotypic and functional characterization of effector cluster of differentiation 8 positive T cells in human patients with primary biliary cirrhosis. Hepatology. 2011;54:1293-1302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 50]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
61.  Yang GX, Wu Y, Tsukamoto H, Leung PS, Lian ZX, Rainbow DB, Hunter KM, Morris GA, Lyons PA, Peterson LB. CD8 T cells mediate direct biliary ductule damage in nonobese diabetic autoimmune biliary disease. J Immunol. 2011;186:1259-1267.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 37]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
62.  Yang GX, Lian ZX, Chuang YH, Moritoki Y, Lan RY, Wakabayashi K, Ansari AA, Flavell RA, Ridgway WM, Coppel RL. Adoptive transfer of CD8(+) T cells from transforming growth factor beta receptor type II (dominant negative form) induces autoimmune cholangitis in mice. Hepatology. 2008;47:1974-1982.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 89]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
63.  Hsu W, Zhang W, Tsuneyama K, Moritoki Y, Ridgway WM, Ansari AA, Coppel RL, Lian ZX, Mackay I, Gershwin ME. Differential mechanisms in the pathogenesis of autoimmune cholangitis versus inflammatory bowel disease in interleukin-2Ralpha(-/-) mice. Hepatology. 2009;49:133-140.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 65]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
64.  Benseler V, Schlitt HJ. [The liver as an immunological organ]. Z Gastroenterol. 2011;49:54-62.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 5]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
65.  Mao TK, Lian ZX, Selmi C, Ichiki Y, Ashwood P, Ansari AA, Coppel RL, Shimoda S, Ishibashi H, Gershwin ME. Altered monocyte responses to defined TLR ligands in patients with primary biliary cirrhosis. Hepatology. 2005;42:802-808.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 137]  [Cited by in F6Publishing: 145]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
66.  Nakamura M, Funami K, Komori A, Yokoyama T, Aiba Y, Araki A, Takii Y, Ito M, Matsuyama M, Koyabu M. Increased expression of Toll-like receptor 3 in intrahepatic biliary epithelial cells at sites of ductular reaction in diseased livers. Hepatol Int. 2008;2:222-230.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 30]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
67.  Zhao J, Zhao S, Zhou G, Liang L, Guo X, Mao P, Zhou X, Wang H, Nan Y, Xu D. Altered biliary epithelial cell and monocyte responses to lipopolysaccharide as a TLR ligand in patients with primary biliary cirrhosis. Scand J Gastroenterol. 2011;46:485-494.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
68.  Wang AP, Migita K, Ito M, Takii Y, Daikoku M, Yokoyama T, Komori A, Nakamura M, Yatsuhashi H, Ishibashi H. Hepatic expression of toll-like receptor 4 in primary biliary cirrhosis. J Autoimmun. 2005;25:85-91.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 74]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
69.  Honda Y, Yamagiwa S, Matsuda Y, Takamura M, Ichida T, Aoyagi Y. Altered expression of TLR homolog RP105 on monocytes hypersensitive to LPS in patients with primary biliary cirrhosis. J Hepatol. 2007;47:404-411.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 31]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
70.  Moritoki Y, Lian ZX, Wulff H, Yang GX, Chuang YH, Lan RY, Ueno Y, Ansari AA, Coppel RL, Mackay IR. AMA production in primary biliary cirrhosis is promoted by the TLR9 ligand CpG and suppressed by potassium channel blockers. Hepatology. 2007;45:314-322.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 67]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
71.  Kikuchi K, Lian ZX, Yang GX, Ansari AA, Ikehara S, Kaplan M, Miyakawa H, Coppel RL, Gershwin ME. Bacterial CpG induces hyper-IgM production in CD27(+) memory B cells in primary biliary cirrhosis. Gastroenterology. 2005;128:304-312.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 103]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
72.  Ambrosini YM, Yang GX, Zhang W, Tsuda M, Shu S, Tsuneyama K, Leung PS, Ansari AA, Coppel RL, Gershwin ME. The multi-hit hypothesis of primary biliary cirrhosis: polyinosinic-polycytidylic acid (poly I: C) and murine autoimmune cholangitis. Clin Exp Immunol. 2011;166:110-120.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 32]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
73.  Kita H, Naidenko OV, Kronenberg M, Ansari AA, Rogers P, He XS, Koning F, Mikayama T, Van De Water J, Coppel RL. Quantitation and phenotypic analysis of natural killer T cells in primary biliary cirrhosis using a human CD1d tetramer. Gastroenterology. 2002;123:1031-1043.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 180]  [Cited by in F6Publishing: 186]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
74.  Chuang YH, Lian ZX, Yang GX, Shu SA, Moritoki Y, Ridgway WM, Ansari AA, Kronenberg M, Flavell RA, Gao B. Natural killer T cells exacerbate liver injury in a transforming growth factor beta receptor II dominant-negative mouse model of primary biliary cirrhosis. Hepatology. 2008;47:571-580.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 95]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
75.  Wu SJ, Yang YH, Tsuneyama K, Leung PS, Illarionov P, Gershwin ME, Chuang YH. Innate immunity and primary biliary cirrhosis: activated invariant natural killer T cells exacerbate murine autoimmune cholangitis and fibrosis. Hepatology. 2011;53:915-925.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 79]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
76.  Shimoda S, Tsuneyama K, Kikuchi K, Harada K, Nakanuma Y, Nakamura M, Ishibashi H, Hisamoto S, Niiro H, Leung PS. The role of natural killer (NK) and NK T cells in the loss of tolerance in murine primary biliary cirrhosis. Clin Exp Immunol. 2012;168:279-284.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
77.  Chuang YH, Lian ZX, Tsuneyama K, Chiang BL, Ansari AA, Coppel RL, Gershwin ME. Increased killing activity and decreased cytokine production in NK cells in patients with primary biliary cirrhosis. J Autoimmun. 2006;26:232-240.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 87]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
78.  Shimoda S, Harada K, Niiro H, Shirabe K, Taketomi A, Maehara Y, Tsuneyama K, Nakanuma Y, Leung P, Ansari AA. Interaction between Toll-like receptors and natural killer cells in the destruction of bile ducts in primary biliary cirrhosis. Hepatology. 2011;53:1270-1281.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 93]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
79.  Oo YH, Shetty S, Adams DH. The role of chemokines in the recruitment of lymphocytes to the liver. Dig Dis. 2010;28:31-44.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 106]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
80.  Chuang YH, Lian ZX, Cheng CM, Lan RY, Yang GX, Moritoki Y, Chiang BL, Ansari AA, Tsuneyama K, Coppel RL. Increased levels of chemokine receptor CXCR3 and chemokines IP-10 and MIG in patients with primary biliary cirrhosis and their first degree relatives. J Autoimmun. 2005;25:126-132.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 72]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
81.  Zhang W, Fei Y, Gao J, Liu B, Zhang F. The role of CXCR3 in the induction of primary biliary cirrhosis. Clin Dev Immunol. 2011;2011:564062.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 10]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
82.  Shimoda S, Harada K, Niiro H, Yoshizumi T, Soejima Y, Taketomi A, Maehara Y, Tsuneyama K, Nakamura M, Komori A. Biliary epithelial cells and primary biliary cirrhosis: the role of liver-infiltrating mononuclear cells. Hepatology. 2008;47:958-965.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 73]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
83.  Shimoda S, Harada K, Niiro H, Taketomi A, Maehara Y, Tsuneyama K, Kikuchi K, Nakanuma Y, Mackay IR, Gershwin ME. CX3CL1 (fractalkine): a signpost for biliary inflammation in primary biliary cirrhosis. Hepatology. 2010;51:567-575.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 89]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
84.  Isse K, Harada K, Zen Y, Kamihira T, Shimoda S, Harada M, Nakanuma Y. Fractalkine and CX3CR1 are involved in the recruitment of intraepithelial lymphocytes of intrahepatic bile ducts. Hepatology. 2005;41:506-516.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 124]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
85.  Aoki CA, Roifman CM, Lian ZX, Bowlus CL, Norman GL, Shoenfeld Y, Mackay IR, Gershwin ME. IL-2 receptor alpha deficiency and features of primary biliary cirrhosis. J Autoimmun. 2006;27:50-53.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 142]  [Cited by in F6Publishing: 136]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
86.  Salas JT, Banales JM, Sarvide S, Recalde S, Ferrer A, Uriarte I, Oude Elferink RP, Prieto J, Medina JF. Ae2a,b-deficient mice develop antimitochondrial antibodies and other features resembling primary biliary cirrhosis. Gastroenterology. 2008;134:1482-1493.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 161]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
87.  Wakabayashi K, Lian ZX, Leung PS, Moritoki Y, Tsuneyama K, Kurth MJ, Lam KS, Yoshida K, Yang GX, Hibi T. Loss of tolerance in C57BL/6 mice to the autoantigen E2 subunit of pyruvate dehydrogenase by a xenobiotic with ensuing biliary ductular disease. Hepatology. 2008;48:531-540.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 126]  [Cited by in F6Publishing: 137]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]