Abstract
Genome-wide association studies (GWAS) have revolutionized the search for genetic influences on complex disorders, such as primary biliary cirrhosis (PBC). Recent GWAS have identified many disease-associated genetic variants. These, overall, highlighted the remarkable contribution of key immunological pathways in PBC that may be involved in the initial mechanisms of loss of tolerance and the subsequent inflammatory response and chronic bile duct damage. Results from GWAS have the potential to be translated in biological knowledge and, hopefully, clinical application. There are a number of immune pathways highlighted in GWAS that may have therapeutic implications in PBC and in other autoimmune diseases, such as the anti-interleukin-12/interleukin-23, nuclear factor-kb, tumor necrosis factor, phosphatidylinositol signaling and hedgehog signaling pathways. Further areas in which GWAS findings are leading to clinical applications either in PBC or in other autoimmune conditions, include disease classification, risk prediction and drug development. In this review we outline the possible next steps that may help accelerate progress from genetic studies to the biological knowledge that would guide the development of predictive, preventive, or therapeutic measures in PBC.
Introduction
Primary biliary cirrhosis (PBC) is the most common autoimmune liver disease and is considered a model of organ-specific autoimmune diseases [1]. It is characterized by loss of tolerance, production of a multilineage immune response to mitochondrial auto-antigens, inflammation of small bile ducts, and in some patients, the development of fibrosis and cirrhosis. Patients with PBC may present with symptoms as fatigue, pruritus and/or jaundice, but the majority of them are asymptomatic at diagnosis. A diagnosis of PBC can be made with confidence in adult patients with otherwise unexplained elevation of alkaline phosphatase and presence of anti-mitochondrial antibodies (AMA) at a titre of ≥1:40 and/or AMA type M2. A liver biopsy is not essential for the diagnosis of PBC in these patients, but allows activity and stage of the disease to be assessed. Progression of disease in PBC is variable with a substantial proportion of patients eventually developing cirrhosis and liver failure. The only licensed therapy for PBC is ursodeoxycholic acid (UDCA) which has been demonstrated to exert anticholestatic effects in various cholestatic disorders. Several potential mechanisms and sites of action of UDCA have been unraveled in clinical and experimental studies which might explain its beneficial effects. These include protection of injured cholangiocytes against the toxic effects of bile acids, particularly at an early stage; stimulation of impaired hepatocellular secretion by mainly post-transcriptional mechanisms, including stimulation of synthesis, targeting and apical membrane insertion of key transporters, more relevant in the advanced cholestasis; stimulation of ductular alkaline choleresis and inhibition of bile acid-induced hepatocyte and cholangiocyte apoptosis.
Many aspects of the basic biology of PBC, including, rigorous definitions of the signature AMA, disease-specific anti-nuclear autoantibodies, the definition of autoreactive CD4+ and CD8+ T-cell responses and the association with some immunological pathways, such as IL-12, NF-κB and TNF, have been elucidated through the development of animal models of PBC, including models that develop fibrosis [2, 3]; and large scale epidemiologic studies, including a number of genome-wide association studies (GWAS) (reviewed in [1, 4–5]). Despite this knowledge, an enormous gap still exists between our knowledge of the etiopathogenesis of PBC and new therapeutic approaches for patients. There has not been a new drug approved for PBC for more than 2 decades and indeed newer biologics merits further investigation to show their safety and efficacy [6]. Since there are a significant number of patients with PBC who do not respond to UDCA [19], there is a strong need for new therapies.
The advent of genome-wide association technology has transformed the landscape of human genetics research. Thanks to GWAS, common genetic variants associated with well-phenotyped diseases, such as inflammatory bowel disease [7] and diabetes [8], have been identified in a non-biased fashion. Such studies are conducted based on the assumption that at least some of the genetic influences on many common diseases are attributable to a limited number of common allelic variants that are present in more than 5% of the population [9] The best-known examples of common disease genes include the ApoE ε4 allele in Alzheimer's disease [10], Factor V (C→A at 1691) allele in deep-venous thrombosis [11], and CKR5Δ32 in resistance to human immunodeficiency virus infection [12].
GWAS typically involve the analysis of hundreds of thousands of common single nucleotide polymorphisms (SNPs) and are not limited to known genes or regulatory regions. These studies require a large sample size not only in order to detect robust associations as false-positive findings arise due to chance alone, but also because of the low effect size of most disease variants detected in GWAS (odds ratios = 1.1−1.4) [13]. The landmark Wellcome Trust Case Control Consortium (WTCCC) study included 2000 cases of each of seven common diseases and 3000 shared controls [14]. It is also mandatory for any GWAS protocol to include a replication of associations claimed to be genuine, in at least one independent case-control panel.
GWAS provide starting points for further biological studies of the affected pathways. Strategies for translating the genetic findings into an applicable understanding of disease pathogenesis are a work in progress. Despite the advent of newer technologies for genetic analysis, in particular sequencing-based methods for identifying disease-associated variants, GWAS-based findings will remain essential, for some time, for designing effective clinical applications. This is in part because the mass of GWAS data that have already been generated continues to be mined for additional trait associations, and because of the unflagging pace with which new GWAS findings are still being published. On the other hand, the prolific associative findings of GWAS are creating a substantial need for downstream genetic, biochemical and cell biological assays to confirm the biological relevance of genotype-phenotype associations and to elucidate the underlying mechanisms of disease.
Recent highlights from high-throughput genetic studies of PBC
In the past decade various GWAS have revealed dozens of disease-associated loci and have provided insights into the allelic architecture of many complex disorders, such as PBC [6–9, 69,70,75]. Large, well-characterized patient cohorts for high-throughput genetic studies of PBC have been established in Europe, North America and Japan; and four GWAS [15–18] and two iCHIP-association [19, 20] studies of PBC have been published.
Similar to the risk alleles identified by GWAS findings for other immune-related conditions, such as rheumatoid arthritis (RA), Crohn's disease (CD) and multiple sclerosis (MS), many of the risk alleles identified in PBC by GWAS are found in conjunction with genes related to immune function, both within and outside the human leukocyte antigen (HLA) [21]. Overall, the data suggest important contributions from a number of immune pathways to the development of PBC (Table 1): from the differentiation of the myeloid cell compartment (SPIB, IRF5, IRF8 and IL-7R) to antigen presentation and T-cell differentiation (IL7R, class II HLA, CD80, IL12, IL12R, TYK2, STAT4, SOCS1) up to B-cell function (SPIB, IRF8, PLC-L2, SPIB, PLC-L2, IKZF3, CXCR5) [22]. Importantly, most of these genes play important roles in many different immune pathways and are not specifically involved in a single, unique function. For instance, IL-7R is induced upon T-cell positive selection and controls thymic CD8 lineage specification and peripheral naive T-cell homeostasis [23] while also having a role in myeloid cell differentiation [24]. Along the same lines, IRF8 is widely involved in immune functions in both innate and adaptive immunity, including B-cell differentiation [25, 26], antigen presentation [27], and homeostasis of the myeloid cell compartment [28].
Table 1.
Locus | SNP | Odd ratio | P-value | Candidate gene | Diseases sharing risk loci with PBCb),c) |
---|---|---|---|---|---|
1p36 [85] | rs3748816 | 1.33 | 3.15E-08 | MMEL1 | - |
1p31.3 [38] | rs72678531 | 1.61 | 2.47E-38 | IL12RB2 | - |
1q31.3 [38] | rs2488393 | 1.28 | 4.29E-12 | DENND1B | CD |
2q32.2 [38] | rs3024921 | 1.62 | 2.59E-18 | STAT4 | CeD, Ra, T1DM, SLE, SSc |
3p24.3 [38] | rs1372072 | 1.2 | 2.28E-08 | PLCL2 | MS |
3q13.3 [38] | rs2293370 | 1.39 | 6.84E-16 | CD80 | MS, CeD, Vit |
3q25.33 [38] | rs2366643 | 1.35 | 3.92E-22 | IL12A | MS, CeD |
4q24 [38] | rs7665090 | 1.26 | 8.48E-14 | NFKB1 | MS,UC |
5p13 [38] | rs6871748 | 1.3 | 2.26E-13 | IL7R | MS, UC |
7p14.1 [38] | rs6974491 | 1.57 | 4.44E-08 | ELMO1 | MS, CeD, PS |
7q32 [38] | rs35188261 | 1.52 | 6.52E-22 | IRF5 | UC, RA, SLE, SSc, |
9q32 [18] | rs4979462 | 1.57 | 1.85E-14 | TNFSF15 | UC, CD |
11q13 [17] | rs538147 | 1.23 | 2.06E-10 | RPS6KA4 | MS, CD, PS, SARC |
11q23.3 [38] | rs80065107 | 1.39 | 7.20E-16 | CXCR5 | MS, CeD, SLE, VIT |
11q23 [38] | rs4938534 | 1.38 | 3.27E-08 | POU2AF1 | CeD |
12p13.2 [38] | rs1800693 | 1.27 | 1.18E-14 | TNFRSF1A | MS CeD, RA, T1DM, VIT, AITD, |
12q24 [38] | rs11065979 | 1.2 | 2.87E-09 | SH2B3 | PSC |
13q14 [10] | rs3862738 | 1.33 | 2.18E-08 | TNFSF11 | CD |
14q24 [38] | rs911263 | 1.26 | 9.95E-11 | RAD51B | - |
14q32 [8] | rs8017161 | 1.22 | 2.61E-13 | TNFAIP2 | - |
16p13.13 [38] | rs12708715 | 1.29 | 2.19E-13 | CLEC16A | MS, UC, T1DM |
16q24.1 [38] | rs11117433 | 1.26 | 1.41E-09 | IRF8 | MS, UC, RA, SSc |
17q12 [38] | rs17564829 | 1.26 | 6.05E-14 | IKZF3 | UC, CD, RA, T1DM |
17q21.1 [38] | rs17564829 | 1.25 | 2.15E-09 | CRHR1 | - |
19p13.2 [38] | rs34536443 | 1.91 | 1.23E-12 | TYK2 | MS, CD, RA, T1DM, SLE, PS |
19q13.3 [38] | rs3745516 | 1.46 | 7.97E-11 | SPIB | - |
22q13.1 [38] | rs2267407 | 1.29 | 1.29E-13 | MAP3K7IP1 | CD |
This table reports risk loci identified at genome-wide level of significance in at least one genome-wide association study or iCHIP study of primary biliary cirrhosis. For each locus, results are from the study with strongest evidence of association.
Phenotypically-associated disorders that share risk loci with PBC are listed.
AITD, autoimmune thyroid disease; CD, Crohn's disease; CeD, celiac disease; T1DM, diabetes mellitus type 1; MS, multiple sclerosis; PS, psoriasis; RA, rheumatoid arthritis; SARC, sarcoidosis; SLE, systemic lupus erythematosis; SSc, systemic sclerosis; UC, ulcerative colitis; VIT, vitiligo.
For most associated loci, there is a substantial lack of understanding regarding the mechanisms by which a genetic variation could influence a phenotype: the identity of the gene(s) affected by the susceptibility variant(s) at each locus is often uncertain, and the mechanisms by which the causal variants (also often unknown) influence phenotype is usually unclear. This uncertainty is a substantial impediment to the understanding needed to make progress towards new therapies or preventive measures. This obstacle highlights the need to pinpoint the causal variants and the genes affected by those variants, as well as the need for informative functional and computational studies to move from gene identification to possible mechanisms that could guide translational progress. As with the translation of many genomic discoveries, translating this work into direct health benefits will require interaction among a wide array of biomedical disciplines, including genomics, molecular biology, clinical medicine, pharmacology and bioinformatics.
Treatment of patients with PBC is not disease-specific due to the lack of knowledge of pathogenic mechanisms. The standard of care is therapy with the secondary bile acid UDCA [29, 30]. The clinical consensus is that a biochemical response to UDCA delays the progression of disease in most patients [31–34]. However, there is a sub-population of patients who do not respond to UDCA and progress more rapidly to liver failure. An inadequate response to UDCA represents a major unmet clinical need in hepatology and GWAS may represent the way forward to address this need.
Areas in which GWAS findings are leading to clinical and therapeutic applications in many diseases include drug development, drug-response studies [35], and risk prediction which can allow patient stratification [62]. Illustrative examples that highlight the potential application of GWAS discoveries in PBC are discussed in some detail below, and other examples on the horizon are briefly listed thereafter.
Possible therapeutic targets highlighted by GWAS
One of the major goals of GWAS findings has been to flag relevant pathways, not previously implicated, in the pathogenesis of complex disorders that could reveal novel therapeutic targets. Explicative findings are the complement pathway in macular degeneration [36] and the autophagy pathway in inflammatory bowel disease [37].
An emerging theme in the genetics of complex disorders is the considerable overlap of genetic susceptibility factors between related diseases. This has been highlighted in the recent primary sclerosing cholangitis (PSC) iCHIP study [38], in which 44 non-HLA loci were correlated in a GWAS of seven clinically associated autoimmune disorders, including ulcerative colitis (UC), CD, T1DM, coeliac disease (CeD), psoriasis, RA and sarcoidosis. In this study, eleven loci of significance were associated at genome-wide level and 33 loci achieved suggestive significance (P<5×10−5) [38]. This suggested a close similarity in the genetic architecture of PSC and each of the other autoimmune conditions. Functional network analysis showed that candidate genes at pleiotropic loci are related in terms of their function, highlighting common pathways involved in the pathogenesis of PSC and other clinically associated disorders. These observations suggest there might be distinct mechanisms by which autoimmunity occurs, each mechanism predisposing to a particular phenotype or set of phenotypes. This might also suggest that there are unique immunologic pathways that should be focussed on for therapeutic intervention. Likewise, in PBC, many of the disease-related variants have been identified in other GWAS of immune-related diseases, with a different mosaic of disease-specific risks contributing to the pathogenesis of PBC. Overall, the data suggest important contributions from a number of immune pathways to the development of PBC. Some of the pathways that may have therapeutic implications in PBC are reported below and in Table 2.
Table 2.
Pathway | Mechanism | Drugs | Autoimmune diseases where modulators (inhibitors) of these pathways have been used in clinical practice |
---|---|---|---|
IL-12/IL-23 | IL-12 promotes Th1-type immune response by driving differentiation of activated, naïve T-cells to Th1 cells. This, together with the interferon-γ (IFN-γ) production IL-12 driven, contributes to loss of tolerance in several models of autoimmunity; IL-23 (which share the p40 subunit of IL-12) is essential for the differentiation of Th17 cells, implicated in the breakdown of immune self-tolerance in PBC |
Anti-IL-12/IL-23 (Ustekinumab) | Psoriasis [44] and Crohn's disease [45, 46] |
NF-κB | Transcription factor which regulates expression of many genes involved in the immune response. NF-κB coinhibitory molecules are key in the prevention of autoimmune diseases. The CD80/CD86-CTLA-4 pathway is the best-characterized inhibitory pathway for T-cell activation | Anti-CD80 (Abatacepi) | Rheumatoid arthritis [35,36] and psoriasis [37] |
TNF-α | Deregulated TNF production characterizes many autoimmune diseases. Recent evidence supports a dualistic, proinflammatory and immune- or diseasesuppressive role for TNF in these conditions | Anti-TNF (Infliximab, Etanercept, Adalimumab, Certolizumab, Golimumab) | ankylosing spondylitis [80], Crohn's disease [81], psosiasis [82], rheumatoid arthritis [83]. |
Phosphatidylinositol signaling | Key pathway for the maintenance of self-tolerance | — | — |
Hedgehog signaling | Pathway involved in the inflammatory and ductular response to cholestatic damage | Hedgehog-inhibitor (Cyclopamine) | Psoriasis [84] |
Anti-interleukin 12 (IL-12)/IL-23
Loci identified in GWAS in PBC suggest a role for T-lymphocyte differentiation in the development of the disease [6–8]. Th1 immune responses have been implicated in many autoimmune diseases [39] and may be involved in the development of autoreactive T cells, consistent with the putative role of the pyruvate dehydrogenase complex (PDC)-specific autoreactive Th1 cells in the pathogenesis of human PBC [40]. Anti-IL-12 signaling promotes Th1-type immune responses by driving differentiation of activated, naïve T cells to Th1 cells. This, together with the IL-12-driven interferon-γ (IFN-γ) production, contributes to loss of tolerance in several models of autoimmunity [41]. Three loci containing genes involved in IL-12 signaling have been identified in GWAS of PBC: the genes IL12A, IL12RB2 [15–17] and STAT4 [17] codifying the subunit p35 of the IL-12, the chain IL12Rβ2 of the IL-12 receptor, and the signal transducer and activator of transcription (STAT4) respectively [42]. Studies conducted in an animal model of PBC have strongly suggested a role for the IL-12 pathway in PBC [43]. Currently, multiple clinical trials have been initiated to test whether monoclonal antibody or transcription-inhibitors of p40 (a subunit of the IL-12 receptor) is of therapeutic benefit in psoriasis [44] and CD [45, 46]. Of note, the p40 subunit of IL-12 is also a component of the dimeric cytokine IL-23, which is essential for the differentiation of Th17 cells. Pilot studies are under way to test the efficacy and safety of the human monoclonal anti-IL-12/IL-23 Ustekinumab in patients with PBC (ClinicalTrials.gov identifier: NCT01389973).
Additional studies are nevertheless required: specifically, genetic association studies and sequencing studies to enable the definition of the specific IL12A and IL12RB2 alleles conferring risk for PBC; molecular analyses that clarify the crosstalk between IL-12 and IL-23 signaling pathways; and in vivo experiments that elucidate the relative contributions of Th17, Treg cells, and other immune cellular subpopulations to PBC. A role for IL-35 is also worthy of investigation, given the subunit nature of the cytokine IL-35 and its receptor, which includes IL-12 p35 and IL-12Rβ2, respectively. Findings from these investigative approaches should then be translated into novel therapy and better outcomes for patients with PBC and other associated autoimmune diseases.
Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB)
Two GWAS in PBC [17, 18] identified loci containing genes involved in activation of nuclear factor κB (NF-κB), a transcription factor which regulates expression of many genes involved in the immune response; NF-κB is also highly activated in other autoimmune disorders such as RA, MS, and asthma [47]. The loci identified in PBC contain the NFKB1 gene itself, and genes in pathways leading to NF-κB activation such as TNFRSF1A, CD80 and RPS6KA4.
Accumulating evidence indicates that coinhibitory molecules, such as CD80, are key in the prevention of autoimmune diseases. The CD80/CD86:CD28/CTLA-4 (cytotoxic T lymphocyte-associated antigen 4) pathway is the best-characterized inhibitory pathway for T-cell activation [48, 49]. CD28 is constitutively expressed on naïve and activated T cells. CD80 is expressed at low levels on resting antigen-presenting cells (APCs) and is upregulated with prolonged interaction with T-cells, whereas CD86 is constitutively expressed and rapidly upregulated on APCs. Thus, CD86 is likely to be mainly involved in mediating initial T-cell activation, while CD80 may play an important role in propagating the immune responses. After activation, T cells express CTLA-4 (CD152). Engagement of CTLA-4 delivers negative signal into T cells, resulting in inhibition and/or termination of T-cell responses.
Taking advantage of the fact that CTLA-4 binds CD80 and CD86 with much higher affinity than CD28 does, a fusion protein consisting of the extracellular domain of CTLA-4 and the constant region of IgG (CTLA-Ig) has been developed to block the interaction between CD80–CD86 and CD28 and thereby inhibit T-cell activation [34]. Such a fusion protein would preferentially inhibit lymphocytes that are in the process of responding to self-antigens without affecting resting T cells that recognize other antigens.
After the encouraging results of in vivo studies in animal models, including PBC models [50], the efficacy of the CTLA-4 Ig (Abatacept) has been examined in patients with autoimmune diseases. Abatacept has shown efficacy in a broad spectrum of RA patients from early stage to refractory diseases that are resistant to TNF blockers [51, 52] and in patients with psoriasis in a phase I trial [53]. Blockade of costimulation between T cells and antigen-presenting cells through CD80 could represent an important therapeutic approach for the treatment of refractory PBC.
Tumor necrosis factor alpha (TNF-α)
TNF-α is an activating factor for a number of intracellular pathways that determine the fate of hepatocytes, and thus plays a key role in liver homeostasis [54]. Interactions between specific members of the TNF pathway lead to the induction of apoptosis as well as the activation of NF-kB signaling, which is anti-apoptotic and pro-inflammatory [55]. GWAS in PBC identified three loci containing genes in TNF-α signaling pathways: TNFRSF1A, DENND1B [17], and TNFAIP2 [17, 18]. TNFRSF1A is one of two receptors for TNF-α; TNFRSF1A−/− mice show attenuated liver fibrosis when compared with wild-type mice after administration of a potent hepatotoxin [56]. DENND1B interacts directly with TNFRSF1A [57] and has previously been associated with asthma [58]. TNF-α signaling also directly induces TNFAIP2 expression [59]. Macrophages from PBC patients, when stimulated with apoptotic bodies from cholangiocytes, produce high levels of TNF-α [60]. Furthermore, serum levels of TNF-α reflect the severity of morphological liver changes in PBC [61]. The TNF signaling pathway was also identified in the pathway analysis of the Italian GWAS cohort [22] where a `self-contained' GWAS pathway analysis method (the linear combination test (LCT)), was applied.
Overall, these findings sustain a prominent role for TNF-α in the pathogenesis of PBC, suggesting that anti-TNF-α treatment, currently used for most inflammatory rheumatic conditions, such as RA, ankylosing spondylitis (AS) and CD, may also represent a promising agent in PBC.
Phosphatidylinositol signaling and hedgehog signaling pathways
Pathway analysis of both the Italian and Canadian GWAS PBC cohorts have highlighted the phosphatidylinositol signaling system pathway, which is an integral component of the adaptive immune response and is essential for the maintenance of self-tolerance [62]. Possible involvement of the phosphatidylinositol pathway in PBC appears to fit well with the TNF hypothesis as this signaling system has been shown to mediate the effects of TNF-α on NF-kB activation [63, 64].
The same pathway analysis also identified the hedgehog (Hh) signaling system, suggesting its involvement in PBC genetic susceptibility. Hh proteins comprise a group of secreted proteins that are involved in organogenesis, and have been shown to promote adult stem cell proliferation [65–67]. Hh signaling has been widely described in PBC. It is involved in the ductular response to cholestatic damage in PBC, characterized by periportal accumulation of proliferating bile ductular cells and associated stromal elements, including myofibroblastic cells and fibrous matrix [68]. Hh signaling was found to be increased in a murine model of bile duct ligation in periportal epithelial cells expressing pan-cytokeratin, representing potential liver progenitor cell populations [53]. Hh signaling has also been shown to be able to promote the survival of biliary epithelial cells, possibly mediated through the inhibition of caspase activity [69]. Lastly, Hh signaling pathway activation has been associated with upregulation of ductular cell expression of genes that promote inflammatory response, such as the gene producing Cxcl16; Hh dependent induction of Cxcl16, demonstrated in both bile duct ligated rats and humans with PBC, resulted in Natural Killer T (NKT) cell chemotaxis towards cholangiocytes in vitro [70].
Hh signaling may represent an important protective factor within the damaged liver, promoting the survival of small periportal epithelial cells representing potential hepatic progenitor cells. Despite the preliminary nature of these studies, the Hh signaling pathway may represent a new therapeutic target to protect or promote cell proliferation and tissue repair within the chronically damaged liver in PBC and other chronic liver diseases.
GWAS in pharmacogenetics
Some scientists believe that, as humans did not evolve in an environment of drug therapies, there is no evolutionary pressure on responses to recently developed pharmacologic agents. Therefore the search for common genetic variants affecting response to pharmacotherapy is likely to be successful because the genetic effect size is likely to be larger than that for disease susceptibility. Since 2007, GWAS have increasingly been applied to pharmacogeneticsto identify loci that affect either drug response or susceptibility to adverse drug reactions. These studies have shown the value of this approach in many fields [71–77].
However, there are limitations in conducting GWAS in pharmacogenetics. First, the variation in drug response is likely to be multifactorial, with many genes working in conjunction with the environment. Second, current GWAS are targeted at elucidating the independent effects of single genes, and may miss interactive or synergistic effects. Furthermore, the challenges in performing adequate replication studies have to be considered for GWAS in pharmacogenetics, particularly when evaluating small cohorts, such as non-responders to UDCA in PBC.
UDCA, which is currently the only available drug in PBC, is thought to work on the downstream events of the pathogenic mechanism of the disease, through reducing the toxicity of bile and reducing bile duct cell apoptosis [78]. There are ongoing studies, focused on exploring, with a GWA approach, the mechanism(s) beyond the lack of biochemical response to UDCA treatment. A major aim of this ongoing project is to identify potential sites for therapeutic intervention in non-responsive patients. New therapeutic targets that may be highlighted by GWAS, as applied to pharmacogenetics, can be localized either in the upstream or downstream processes of PBC pathogenesis; from the mechanisms that lead to loss of tolerance to the fibrotic phase secondary to cholestasis. Furthermore, improved knowledge of the genetic basis of the lack of response to UDCA will allow to identify non-responders at an early stage and to select them for next-generation drug trials.
Risk prediction
Attempting to predict the onset and progression of disease is one of the cornerstones of epidemiology. GWAS show significant potential to identify molecular factors that enable patient stratification and might prove useful in personalized medicine. Accurate risk prediction can enable targeted preventative treatments or more intensive follow-up, particularly for patients at high risk of progression.
The success of recent GWAS has rapidly changed the outlook for genetic risk prediction. These studies have unlocked thousands of clearly validated genetic associations to complex diseases, but their generally weak effects have left their predictive value and clinical utility subject to hot debate. GWAS data might find ready application in risk prediction in PBC in those patients identified at an early stage of the disease. Risk stratification at an early stage may be important from the perspective of developing treatments that either prevent disease entirely or that improve the outcome when instituted before biliary fibrosis and cirrhosis develop.
Progression of disease in PBC is variable with a substantial proportion of patients eventually developing cirrhosis and liver failure, while other patients remain well and never develop liver-related problems. Progression of disease may represent a complex trait with genetics factors and environmental factors playing together. Genetic variants associated with disease progression detected with GWAS can allow identifying patients at high risk of progressive disease for whom second-line “targeted” therapies would be a valuable therapeutic option. Studies aiming to identify common genetic variants associated with disease progression in PBC at genome-wide level of significance, are currently in progress. It is unlikely that genetic variants associated with disease progression are similar to those associated with susceptibility to PBC. More likely, these studies will identify genetic variants associated with fibrosis progression, which may be then extrapolated for other liver diseases and translated into clinical practice.
Predictive accuracy from genetic models varies greatly across diseases, but the range is similar to that of non-genetic risk-prediction models. A significant improvement in reclassification statistics compared to established clinical risk factors alone is possible. In a cohort that had been classified for risk of cardiovascular events, a combination of genetic variants associated with cholesterol levels was used to develop a genotype score for reclassification [79]. As a result, of the 26% of the study cohort that had been initially estimated to be at intermediate risk, 35% (9% of the total cohort) were reclassified into low- or high-risk categories [79]. For PBC, where non-genetic prediction of outcome has already been explored in preliminary studies with the use of the liver function tests at presentation, it is important to evaluate the information added by genetic loci. Clearly, if classical prediction is strong and genetic prediction is weak, little additional value is added. Furthermore, GWAS risk factors are not necessarily independent of the classical predictors.
There are a number of benefits of such genetic prediction over classical alternatives. For instance, unlike classical clinical risk prediction, genetic risk prediction is highly stable over time, as a person's genetic sequence is essentially constant throughout their life. Such stable risk stratification could be especially important when the proposed interventions are more effective if started at an early age, or continued over a long time period. The utility of genetic risk prediction is dependent not just on predictive accuracy, but also on cost and the ability of clinicians and patients to effectively use this information. The falling cost of whole-genome sequencing will drive the marginal cost of prediction lower, but further progress in gene-mapping research, infrastructure and medical practice will be needed to take full advantage of genetic risk prediction.
Conclusion
During the past decade, there have been significant advances in our understanding of the immunobiology of PBC and, in particular, a rigorous dissection of not only the serologic abnormalities, including AMA, but also the definition of autoreactive CD4+ and CD8+ T cells. Further, there is increasing evidence for the interplay of genetic and environmental factors in individual host susceptibility. Prior to the advent of GWAS, only class II HLA loci had been reproducibly shown to associate with disease [21]. Non-HLA loci were suggested for several genes (e.g., CTLA-4, MDR3), but often inconclusively replicated. With the application of genome-wide technology, HLA was confirmed as the strongest association and many other risk loci have been identified, with equivalent effect size to HLA, including IL12A, IL12RB2, STAT4, IRF5-TNPO3, 17q12.21, MMEL1, SPIB, and CTLA-4. Pathways such as TNF signaling, antigen processing and presentation, and apoptosis, each of which is an established contributor to genetic predisposition to PBC, are among the top pathways identified trough GWAS. These studies highlight the interplay between innate and acquired immunity in PBC. Elucidating the effects of these pathways in PBC is complicated, and it will require additional studies that clarify the effector mechanisms involved; indeed response to therapy, clinical progression, and symptoms remain additional areas for further dedicated studies, and in which different genetic risk factors may be relevant. Nowadays, identification of risk loci associated with disease is leading to the development of rational, disease-specific, therapies for the future.
Acknowledgements
MC is supported in part by the Dame Sheila Sherlock EASL Fellowship Program of the European association for the study of the liver (EASL). AL and PI are supported in part by the the National Institute of Health (N.I.H.) grant #DK091823-01A1.
Footnotes
Conflict of Interest The authors declare no financial or commercial conflict of interest.
BIBLIOGRAPHY
- 1.Invernizzi P, Selmi C, Gershwin ME. Update on primary biliary cirrhosis. Dig Liver Dis. 2010;42:401–408. doi: 10.1016/j.dld.2010.02.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Tsuda M, Zhang W, Yang GX, Tsuneyama K, Ando Y, Kawata K, Park O, et al. Deletion of interleukin (IL)-12p35 induces liver fibrosis in dominant-negative TGFbeta receptor type II mice. Hepatology. 2013;57:806–816. doi: 10.1002/hep.25829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Ando Y, Yang GX, Tsuda M, Kawata K, Zhang W, Nakajima T, Tsuneyama K, et al. The immunobiology of colitis and cholangitis in interleukin-23p19 and interleukin-17A deleted dominant negative form of transforming growth factor beta receptor type II mice. Hepatology. 2012;56:1418–1426. doi: 10.1002/hep.25803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Lleo A, Invernizzi P. Apotopes and innate immune system: novel players in the primary biliary cirrhosis scenario. Dig Liver Dis. 2013;45:630–636. doi: 10.1016/j.dld.2013.01.005. [DOI] [PubMed] [Google Scholar]
- 5.Podda M, Selmi C, Lleo A, Moroni L, Invernizzi P. The limitations and hidden gems of the epidemiology of primary biliary cirrhosis. J Autoimmun. 2013;46:81–87. doi: 10.1016/j.jaut.2013.06.015. [DOI] [PubMed] [Google Scholar]
- 6.Tsuda M, Moritoki Y, Lian ZX, Zhang W, Yoshida K, Wakabayashi K, Yang GX, et al. Biochemical and immunologic effects of rituximab in patients with primary biliary cirrhosis and an incomplete response to ursodeoxycholic acid. Hepatology. 2012;55:512–521. doi: 10.1002/hep.24748. [DOI] [PubMed] [Google Scholar]
- 7.Duerr RH, Taylor KD, Brant SR, Rioux JD, Silverberg MS, Daly MJ, Steinhart AH, et al. A genome-wide association study identifies IL23R as an inflammatory bowel disease gene. Science. 2006;314:1461–1463. doi: 10.1126/science.1135245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Sladek R, Rocheleau G, Rung J, Dina C, Shen L, Serre D, Boutin P, et al. A genome-wide association study identifies novel risk loci for type 2 diabetes. Nature. 2007;445:881–885. doi: 10.1038/nature05616. [DOI] [PubMed] [Google Scholar]
- 9.Collins FS, Guyer MS, Charkravarti A. Variations on a theme: cataloging human DNA sequence variation. Science. 1997;278:1580–1581. doi: 10.1126/science.278.5343.1580. [DOI] [PubMed] [Google Scholar]
- 10.Saunders AM, Strittmatter WJ, Schmechel D, George-Hyslop PH, Pericak-Vance MA, Joo SH, Rosi BL, et al. Association of apolipoprotein E allele epsilon 4 with late-onset familial and sporadic Alzheimer's disease. Neurology. 1993;43:1467–1472. doi: 10.1212/wnl.43.8.1467. [DOI] [PubMed] [Google Scholar]
- 11.Bertina RM, Koeleman BP, Koster T, Rosendaal FR, Dirven RJ, de Ronde H, van der Velden PA, et al. Mutation in blood coagulation factor V associated with resistance to activated protein C. Nature. 1994;369:64–67. doi: 10.1038/369064a0. [DOI] [PubMed] [Google Scholar]
- 12.Dean M, Carrington M, Winkler C, Huttley GA, Smith MW, Allikmets R, Goedert JJ, et al. Genetic restriction of HIV-1 infection and progression to AIDS by a deletion allele of the CKR5 structural gene. Hemophilia Growth and Development Study, Multicenter AIDS Cohort Study, Multicenter Hemophilia Cohort Study, San Francisco City Cohort, ALIVE Study. Science. 1996;273:1856–1862. doi: 10.1126/science.273.5283.1856. [DOI] [PubMed] [Google Scholar]
- 13.Hindorff LA, Sethupathy P, Junkins HA, Ramos EM, Mehta JP, Collins FS, Manolio TA. Potential etiologic and functional implications of genome-wide association loci for human diseases and traits. Proc Natl Acad Sci U S A. 2009;106:9362–9367. doi: 10.1073/pnas.0903103106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Wellcome Trust Case Control, C Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature. 2007;447:661–678. doi: 10.1038/nature05911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Hirschfield GM, Liu X, Xu C, Lu Y, Xie G, Lu Y, Gu X, et al. Primary biliary cirrhosis associated with HLA, IL12A, and IL12RB2 variants. N Engl J Med. 2009;360:2544–2555. doi: 10.1056/NEJMoa0810440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Liu X, Invernizzi P, Lu Y, Kosoy R, Lu Y, Bianchi I, Podda M, et al. Genome-wide meta-analyses identify three loci associated with primary biliary cirrhosis. Nat Genet. 2010;42:658–660. doi: 10.1038/ng.627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Mells GF, Floyd JA, Morley KI, Cordell HJ, Franklin CS, Shin SY, Heneghan MA, et al. Genome-wide association study identifies 12 new susceptibility loci for primary biliary cirrhosis. Nat Genet. 2011;43:329–332. doi: 10.1038/ng.789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Nakamura M, Nishida N, Kawashima M, Aiba Y, Tanaka A, Yasunami M, Nakamura H, et al. Genome-wide association study identifies TNFSF15 and POU2AF1 as susceptibility loci for primary biliary cirrhosis in the Japanese population. Am J Hum Genet. 2012;91:721–728. doi: 10.1016/j.ajhg.2012.08.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Juran BD, Hirschfield GM, Invernizzi P, Atkinson EJ, Li Y, Xie G, Kosoy R, et al. Immunochip analyses identify a novel risk locus for primary biliary cirrhosis at 13q14, multiple independent associations at four established risk loci and epistasis between 1p31 and 7q32 risk variants. Hum Mol Genet. 2012;21:5209–5221. doi: 10.1093/hmg/dds359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Liu JZ, Almarri MA, Gaffney DJ, Mells GF, Jostins L, Cordell HJ, Ducker SJ, et al. Dense fine-mapping study identifies new susceptibility loci for primary biliary cirrhosis. Nat Genet. 2012;44:1137–1141. doi: 10.1038/ng.2395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Invernizzi P. Human leukocyte antigen in primary biliary cirrhosis: an old story now reviving. Hepatology. 2011;54:714–723. doi: 10.1002/hep.24414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Kar SP, Seldin MF, Chen W, Lu E, Hirschfield GM, Invernizzi P, Heathcote J, et al. Pathway-based analysis of primary biliary cirrhosis genome-wide association studies. Genes Immun. 2013;14:179–186. doi: 10.1038/gene.2013.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Ouyang W, Oh SA, Ma Q, Bivona MR, Zhu J, Li MO. TGF-beta cytokine signaling promotes CD8+ T cell development and low-affinity CD4+ T cell homeostasis by regulation of interleukin-7 receptor alpha expression. Immunity. 2013;39:335–346. doi: 10.1016/j.immuni.2013.07.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.McKay FC, Hoe E, Parnell G, Gatt P, Schibeci SD, Stewart GJ, Booth DR. IL7Ralpha Expression and Upregulation by IFNbeta in Dendritic Cell Subsets Is Haplotype-Dependent. PLoS One. 2013;8:e77508. doi: 10.1371/journal.pone.0077508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Tarte K, Zhan F, De Vos J, Klein B, Shaughnessy J., Jr. Gene expression profiling of plasma cells and plasmablasts: toward a better understanding of the late stages of B-cell differentiation. Blood. 2003;102:592–600. doi: 10.1182/blood-2002-10-3161. [DOI] [PubMed] [Google Scholar]
- 26.Zhan F, Tian E, Bumm K, Smith R, Barlogie B, Shaughnessy J., Jr. Gene expression profiling of human plasma cell differentiation and classification of multiple myeloma based on similarities to distinct stages of late-stage B-cell development. Blood. 2003;101:1128–1140. doi: 10.1182/blood-2002-06-1737. [DOI] [PubMed] [Google Scholar]
- 27.Shin DM, Lee CH, Morse HC., 3rd IRF8 governs expression of genes involved in innate and adaptive immunity in human and mouse germinal center B cells. PLoS One. 2011;6:e27384. doi: 10.1371/journal.pone.0027384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Waight JD, Netherby C, Hensen ML, Miller A, Hu Q, Liu S, Bogner PN, et al. Myeloid-derived suppressor cell development is regulated by a STAT/IRF-8 axis. J Clin Invest. 2013;123:4464–4478. doi: 10.1172/JCI68189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Poupon RE, Balkau B, Eschwege E, Poupon R. A multicenter, controlled trial of ursodiol for the treatment of primary biliary cirrhosis. UDCA-PBC Study Group. N Engl J Med. 1991;324:1548–1554. doi: 10.1056/NEJM199105303242204. [DOI] [PubMed] [Google Scholar]
- 30.Poupon RE, Poupon R, Balkau B. Ursodiol for the long-term treatment of primary biliary cirrhosis. The UDCA-PBC Study Group. N Engl J Med. 1994;330:1342–1347. doi: 10.1056/NEJM199405123301903. [DOI] [PubMed] [Google Scholar]
- 31.Kuiper EM, Hansen BE, de Vries RA, den Ouden-Muller JW, van Ditzhuijsen TJ, Haagsma EB, Houben MH, et al. Improved prognosis of patients with primary biliary cirrhosis that have a biochemical response to ursodeoxycholic acid. Gastroenterology. 2009;136:1281–1287. doi: 10.1053/j.gastro.2009.01.003. [DOI] [PubMed] [Google Scholar]
- 32.Corpechot C, Abenavoli L, Rabahi N, Chretien Y, Andreani T, Johanet C, Chazouilleres O, et al. Biochemical response to ursodeoxycholic acid and long-term prognosis in primary biliary cirrhosis. Hepatology. 2008;48:871–877. doi: 10.1002/hep.22428. [DOI] [PubMed] [Google Scholar]
- 33.Pares A, Caballeria L, Rodes J. Excellent long-term survival in patients with primary biliary cirrhosis and biochemical response to ursodeoxycholic Acid. Gastroenterology. 2006;130:715–720. doi: 10.1053/j.gastro.2005.12.029. [DOI] [PubMed] [Google Scholar]
- 34.Carbone M, Mells GF, Pells G, Dawwas MF, Newton JL, Heneghan MA, Neuberger JM, et al. Sex and age are determinants of the clinical phenotype of primary biliary cirrhosis and response to ursodeoxycholic acid. Gastroenterology. 2013;144:560–569. e567. doi: 10.1053/j.gastro.2012.12.005. quiz e513–564. [DOI] [PubMed] [Google Scholar]
- 35.Fellay J, Thompson AJ, Ge D, Gumbs CE, Urban TJ, Shianna KV, Little LD, et al. ITPA gene variants protect against anaemia in patients treated for chronic hepatitis C. Nature. 2010;464:405–408. doi: 10.1038/nature08825. [DOI] [PubMed] [Google Scholar]
- 36.Klein RJ, Zeiss C, Chew EY, Tsai JY, Sackler RS, Haynes C, Henning AK, et al. Complement factor H polymorphism in age-related macular degeneration. Science. 2005;308:385–389. doi: 10.1126/science.1109557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Rioux JD, Xavier RJ, Taylor KD, Silverberg MS, Goyette P, Huett A, Green T, et al. Genome-wide association study identifies new susceptibility loci for Crohn disease and implicates autophagy in disease pathogenesis. Nat Genet. 2007;39:596–604. doi: 10.1038/ng2032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Liu JZ, Hov JR, Folseraas T, Ellinghaus E, Rushbrook SM, Doncheva NT, Andreassen OA, et al. Dense genotyping of immune-related disease regions identifies nine new risk loci for primary sclerosing cholangitis. Nat Genet. 2013;45:670–675. doi: 10.1038/ng.2616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Zhernakova A, van Diemen CC, Wijmenga C. Detecting shared pathogenesis from the shared genetics of immune-related diseases. Nat Rev Genet. 2009;10:43–55. doi: 10.1038/nrg2489. [DOI] [PubMed] [Google Scholar]
- 40.Kita H, Matsumura S, He XS, Ansari AA, Lian ZX, Van de Water J, Coppel RL, et al. Quantitative and functional analysis of PDC-E2-specific autoreactive cytotoxic T lymphocytes in primary biliary cirrhosis. J Clin Invest. 2002;109:1231–1240. doi: 10.1172/JCI14698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Becher B, Durell BG, Noelle RJ. Experimental autoimmune encephalitis and inflammation in the absence of interleukin-12. J Clin Invest. 2002;110:493–497. doi: 10.1172/JCI15751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Lleo A, Gershwin ME, Mantovani A, Invernizzi P. Towards common denominators in primary biliary cirrhosis: the role of IL-12. J Hepatol. 2012;56:731–733. doi: 10.1016/j.jhep.2011.05.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Yoshida K, Yang GX, Zhang W, Tsuda M, Tsuneyama K, Moritoki Y, Ansari AA, et al. Deletion of interleukin-12p40 suppresses autoimmune cholangitis in dominant negative transforming growth factor beta receptor type II mice. Hepatology. 2009;50:1494–1500. doi: 10.1002/hep.23132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Leonardi CL, Kimball AB, Papp KA, Yeilding N, Guzzo C, Wang Y, Li S, et al. Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 76-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 1) Lancet. 2008;371:1665–1674. doi: 10.1016/S0140-6736(08)60725-4. [DOI] [PubMed] [Google Scholar]
- 45.Mannon PJ, Fuss IJ, Mayer L, Elson CO, Sandborn WJ, Present D, Dolin B, et al. Anti-interleukin-12 antibody for active Crohn's disease. N Engl J Med. 2004;351:2069–2079. doi: 10.1056/NEJMoa033402. [DOI] [PubMed] [Google Scholar]
- 46.Burakoff R, Barish CF, Riff D, Pruitt R, Chey WY, Farraye FA, Shafran I, et al. A phase 1/2A trial of STA 5326, an oral interleukin-12/23 inhibitor, in patients with active moderate to severe Crohn's disease. Inflamm Bowel Dis. 2006;12:558–565. doi: 10.1097/01.ibd.0000225337.14356.31. [DOI] [PubMed] [Google Scholar]
- 47.Li Q, Verma IM. NF-kappaB regulation in the immune system. Nat Rev Immunol. 2002;2:725–734. doi: 10.1038/nri910. [DOI] [PubMed] [Google Scholar]
- 48.Greenwald RJ, Freeman GJ, Sharpe AH. The B7 family revisited. Annu Rev Immunol. 2005;23:515–548. doi: 10.1146/annurev.immunol.23.021704.115611. [DOI] [PubMed] [Google Scholar]
- 49.Scalapino KJ, Daikh DI. CTLA-4: a key regulatory point in the control of autoimmune disease. Immunol Rev. 2008;223:143–155. doi: 10.1111/j.1600-065X.2008.00639.x. [DOI] [PubMed] [Google Scholar]
- 50.Dhirapong A, Yang GX, Nadler S, Zhang W, Tsuneyama K, Leung P, Knechtle S, et al. Therapeutic effect of cytotoxic T lymphocyte antigen 4/immunoglobulin on a murine model of primary biliary cirrhosis. Hepatology. 2013;57:708–715. doi: 10.1002/hep.26067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Genovese MC, Becker JC, Schiff M, Luggen M, Sherrer Y, Kremer J, Birbara C, et al. Abatacept for rheumatoid arthritis refractory to tumor necrosis factor alpha inhibition. N Engl J Med. 2005;353:1114–1123. doi: 10.1056/NEJMoa050524. [DOI] [PubMed] [Google Scholar]
- 52.Kremer JM, Dougados M, Emery P, Durez P, Sibilia J, Shergy W, Steinfeld S, et al. Treatment of rheumatoid arthritis with the selective costimulation modulator abatacept: twelve-month results of a phase iib, double-blind, randomized, placebo-controlled trial. Arthritis Rheum. 2005;52:2263–2271. doi: 10.1002/art.21201. [DOI] [PubMed] [Google Scholar]
- 53.Abrams JR, Lebwohl MG, Guzzo CA, Jegasothy BV, Goldfarb MT, Goffe BS, Menter A, et al. CTLA4Ig-mediated blockade of T-cell costimulation in patients with psoriasis vulgaris. J Clin Invest. 1999;103:1243–1252. doi: 10.1172/JCI5857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Tacke F, Luedde T, Trautwein C. Inflammatory pathways in liver homeostasis and liver injury. Clin Rev Allergy Immunol. 2009;36:4–12. doi: 10.1007/s12016-008-8091-0. [DOI] [PubMed] [Google Scholar]
- 55.Locksley RM, Killeen N, Lenardo MJ. The TNF and TNF receptor superfamilies: integrating mammalian biology. Cell. 2001;104:487–501. doi: 10.1016/s0092-8674(01)00237-9. [DOI] [PubMed] [Google Scholar]
- 56.Kitamura K, Nakamoto Y, Akiyama M, Fujii C, Kondo T, Kobayashi K, Kaneko S, et al. Pathogenic roles of tumor necrosis factor receptor p55-mediated signals in dimethylnitrosamine-induced murine liver fibrosis. Lab Invest. 2002;82:571–583. doi: 10.1038/labinvest.3780452. [DOI] [PubMed] [Google Scholar]
- 57.Del Villar K, Miller CA. Down-regulation of DENN/MADD, a TNF receptor binding protein, correlates with neuronal cell death in Alzheimer's disease brain and hippocampal neurons. Proc Natl Acad Sci U S A. 2004;101:4210–4215. doi: 10.1073/pnas.0307349101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Sleiman PM, Flory J, Imielinski M, Bradfield JP, Annaiah K, Willis-Owen SA, Wang K, et al. Variants of DENND1B associated with asthma in children. N Engl J Med. 2010;362:36–44. doi: 10.1056/NEJMoa0901867. [DOI] [PubMed] [Google Scholar]
- 59.Sarma V, Wolf FW, Marks RM, Shows TB, Dixit VM. Cloning of a novel tumor necrosis factor-alpha-inducible primary response gene that is differentially expressed in development and capillary tube-like formation in vitro. J Immunol. 1992;148:3302–3312. [PubMed] [Google Scholar]
- 60.Lleo A, Bowlus CL, Yang GX, Invernizzi P, Podda M, Van de Water J, Ansari AA, et al. Biliary apotopes and anti-mitochondrial antibodies activate innate immune responses in primary biliary cirrhosis. Hepatology. 2010;52:987–998. doi: 10.1002/hep.23783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Neuman M, Angulo P, Malkiewicz I, Jorgensen R, Shear N, Dickson ER, Haber J, et al. Tumor necrosis factor-alpha and transforming growth factor-beta reflect severity of liver damage in primary biliary cirrhosis. J Gastroenterol Hepatol. 2002;17:196–202. doi: 10.1046/j.1440-1746.2002.02672.x. [DOI] [PubMed] [Google Scholar]
- 62.Koyasu S. The role of PI3K in immune cells. Nat Immunol. 2003;4:313–319. doi: 10.1038/ni0403-313. [DOI] [PubMed] [Google Scholar]
- 63.Hochdorfer T, Kuhny M, Zorn CN, Hendriks RW, Vanhaesebroeck B, Bohnacker T, Krystal G, et al. Activation of the PI3K pathway increases TLR-induced TNF-alpha and IL-6 but reduces IL-1beta production in mast cells. Cell Signal. 2011;23:866–875. doi: 10.1016/j.cellsig.2011.01.012. [DOI] [PubMed] [Google Scholar]
- 64.Frey RS, Gao X, Javaid K, Siddiqui SS, Rahman A, Malik AB. Phosphatidylinositol 3-kinase gamma signaling through protein kinase Czeta induces NADPH oxidase-mediated oxidant generation and NF-kappaB activation in endothelial cells. J Biol Chem. 2006;281:16128–16138. doi: 10.1074/jbc.M508810200. [DOI] [PubMed] [Google Scholar]
- 65.Varjosalo M, Taipale J. Hedgehog: functions and mechanisms. Genes Dev. 2008;22:2454–2472. doi: 10.1101/gad.1693608. [DOI] [PubMed] [Google Scholar]
- 66.Ingham PW, McMahon AP. Hedgehog signaling in animal development: paradigms and principles. Genes Dev. 2001;15:3059–3087. doi: 10.1101/gad.938601. [DOI] [PubMed] [Google Scholar]
- 67.Bhardwaj G, Murdoch B, Wu D, Baker DP, Williams KP, Chadwick K, Ling LE, et al. Sonic hedgehog induces the proliferation of primitive human hematopoietic cells via BMP regulation. Nat Immunol. 2001;2:172–180. doi: 10.1038/84282. [DOI] [PubMed] [Google Scholar]
- 68.Jung Y, McCall SJ, Li YX, Diehl AM. Bile ductules and stromal cells express hedgehog ligands and/or hedgehog target genes in primary biliary cirrhosis. Hepatology. 2007;45:1091–1096. doi: 10.1002/hep.21660. [DOI] [PubMed] [Google Scholar]
- 69.Omenetti A, Popov Y, Jung Y, Choi SS, Witek RP, Yang L, Brown KD, et al. The hedgehog pathway regulates remodelling responses to biliary obstruction in rats. Gut. 2008;57:1275–1282. doi: 10.1136/gut.2008.148619. [DOI] [PubMed] [Google Scholar]
- 70.Omenetti A, Syn WK, Jung Y, Francis H, Porrello A, Witek RP, Choi SS, et al. Repair-related activation of hedgehog signaling promotes cholangiocyte chemokine production. Hepatology. 2009;50:518–527. doi: 10.1002/hep.23019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Ge D, Fellay J, Thompson AJ, Simon JS, Shianna KV, Urban TJ, Heinzen EL, et al. Genetic variation in IL28B predicts hepatitis C treatment-induced viral clearance. Nature. 2009;461:399–401. doi: 10.1038/nature08309. [DOI] [PubMed] [Google Scholar]
- 72.Suppiah V, Moldovan M, Ahlenstiel G, Berg T, Weltman M, Abate ML, Bassendine M, et al. IL28B is associated with response to chronic hepatitis C interferon-alpha and ribavirin therapy. Nat Genet. 2009;41:1100–1104. doi: 10.1038/ng.447. [DOI] [PubMed] [Google Scholar]
- 73.Tanaka Y, Nishida N, Sugiyama M, Kurosaki M, Matsuura K, Sakamoto N, Nakagawa M, et al. Genome-wide association of IL28B with response to pegylated interferon-alpha and ribavirin therapy for chronic hepatitis C. Nat Genet. 2009;41:1105–1109. doi: 10.1038/ng.449. [DOI] [PubMed] [Google Scholar]
- 74.Shuldiner AR, O'Connell JR, Bliden KP, Gandhi A, Ryan K, Horenstein RB, Damcott CM, et al. Association of cytochrome P450 2C19 genotype with the antiplatelet effect and clinical efficacy of clopidogrel therapy. JAMA. 2009;302:849–857. doi: 10.1001/jama.2009.1232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Cooper GM, Johnson JA, Langaee TY, Feng H, Stanaway IB, Schwarz UI, Ritchie MD, et al. A genome-wide scan for common genetic variants with a large influence on warfarin maintenance dose. Blood. 2008;112:1022–1027. doi: 10.1182/blood-2008-01-134247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Takeuchi F, McGinnis R, Bourgeois S, Barnes C, Eriksson N, Soranzo N, Whittaker P, et al. A genome-wide association study confirms VKORC1, CYP2C9, and CYP4F2 as principal genetic determinants of warfarin dose. PLoS Genet. 2009;5:e1000433. doi: 10.1371/journal.pgen.1000433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Teichert M, Eijgelsheim M, Rivadeneira F, Uitterlinden AG, van Schaik RH, Hofman A, De Smet PA, et al. A genome-wide association study of acenocoumarol maintenance dosage. Hum Mol Genet. 2009;18:3758–3768. doi: 10.1093/hmg/ddp309. [DOI] [PubMed] [Google Scholar]
- 78.Paumgartner G, Beuers U. Mechanisms of action and therapeutic efficacy of ursodeoxycholic acid in cholestatic liver disease. Clin Liver Dis. 2004;8:67–81. vi. doi: 10.1016/S1089-3261(03)00135-1. [DOI] [PubMed] [Google Scholar]
- 79.Kathiresan S, Melander O, Anevski D, Guiducci C, Burtt NP, Roos C, Hirschhorn JN, et al. Polymorphisms associated with cholesterol and risk of cardiovascular events. N Engl J Med. 2008;358:1240–1249. doi: 10.1056/NEJMoa0706728. [DOI] [PubMed] [Google Scholar]
- 80.Braun J, Davis J, Dougados M, Sieper J, van der Linden S, van der Heijde D, Group AW. First update of the international ASAS consensus statement for the use of anti-TNF agents in patients with ankylosing spondylitis. Ann Rheum Dis. 2006;65:316–320. doi: 10.1136/ard.2005.040758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Rutgeerts P, Vermeire S, Van Assche G. Biological therapies for inflammatory bowel diseases. Gastroenterology. 2009;136:1182–1197. doi: 10.1053/j.gastro.2009.02.001. [DOI] [PubMed] [Google Scholar]
- 82.Gisondi P, Girolomoni G. Biologic therapies in psoriasis: a new therapeutic approach. Autoimmun Rev. 2007;6:515–519. doi: 10.1016/j.autrev.2006.12.002. [DOI] [PubMed] [Google Scholar]
- 83.Feldmann M, Maini RN. Lasker Clinical Medical Research Award. TNF defined as a therapeutic target for rheumatoid arthritis and other autoimmune diseases. Nat Med. 2003;9:1245–1250. doi: 10.1038/nm939. [DOI] [PubMed] [Google Scholar]
- 84.Tas S, Avci O. Rapid clearance of psoriatic skin lesions induced by topical cyclopamine. A preliminary proof of concept study. Dermatology. 2004;209:126–131. doi: 10.1159/000079596. [DOI] [PubMed] [Google Scholar]
- 85.Hirschfield GM, Liu X, Han Y, Gorlov IP, Lu Y, Xu C, Lu Y, et al. Variants at IRF5-TNPO3, 17q12-21 and MMEL1 are associated with primary biliary cirrhosis. Nat Genet. 2010;42:655–657. doi: 10.1038/ng.631. [DOI] [PMC free article] [PubMed] [Google Scholar]