Skip to main content
  • Review Article
  • Published:

The X-factor in primary biliary cirrhosis: monosomy X and xenobiotics

Abstract

Primary biliary cirrhosis (PBC) is a chronic, cholestatic, autoimmune liver disease characterised by the destruction of small- and medium-sized bile ducts. The serological hallmark of PBC includes antimitochondrial antibodies (AMA). The disease has a striking female predominance, and primarily affects women of middle-age. First-degree relatives, and in particular female relatives, are known to have an increased risk of developing the disease. Several studies have attempted to explain the female predominance of PBC, and autoimmune diseases in general. Two components that are of interest in PBC include monosomy X and xenobiotics. Monosomy X has been noted to be prevalent in the peripheral blood mononuclear cells of PBC patients. Xenobiotics, which are exogenous chemicals not normally found within the body, have been implicated in the modification of, and loss of, tolerance to AMA. Several cosmetics are known to contain these xenobiotics, which is of interest given the information provided in regards to known risk factors for PBC development. This review will focus on X monosomy and xenobiotics, which appear to constitute the X-factor of PBC.

Introduction

Primary biliary cirrhosis (PBC) is a chronic cholestatic liver disease with characteristic immune-mediated destruction of the small- and medium-sized intrahepatic bile ducts, with progression of fibrosis to cirrhosis and liver failure, at which time transplantation is required [1–5]. At the time of diagnosis, patients may be symptomatic or asymptomatic. In asymptomatic cases, patients may have normal or abnormal biochemistry tests, with cholestatic indices being raised [1–3, 6–9]. Symptomatology of PBC generally includes non-specific symptoms such as fatigue, pruritus and arthralgias, with liver disease not being suspected initially [1–3, 6–9]. Advanced symptoms may be related to portal hypertension and hepatic decompensation, including jaundice, ascites or variceal bleeding [1–3, 6–9]. Both symptomatic and asymptomatic patients are usually seropositive for disease-specific autoantibodies such as anti-mitochondrial (AMA) or disease-specific antinuclear antibody (ANA) [10–14]. The diagnostic criterion of PBC includes: biochemical evidence of cholestasis, the presence of disease-specific AMA and/or ANA, and PBC-specific histopathology [2, 8, 9]. PBC-specific histopathological features include biliary epithelial cell destruction, ductopaenia, portal inflammatory cell infiltration and granuloma formation [2, 3]. Raised alkaline phosphatase and γGT are indicative of cholestasis [1–3, 6–9].

AMA are present in up to 95 % of PBC patients, and are indicative of future development of PBC in asymptomatic patients [1–3, 6–12, 15–17]. This is in contrast to the general population, where the prevalence of AMA is <1 % [18–20]. PBC-specific AMA are directed against components of the 2-oxo-acid dehydrogenase complexes (previously known as M2 antigens), but predominantly recognise the E2 subunit of the pyruvate dehydrogenase complex (PDC) [1–3, 12, 15–17, 21–26]. AMA-positive cases with PBC have antibodies against PDC-E2 in 90 % of cases, and these antibodies also cross-react with the PDC-E3 binding protein (E3BP) [27–29]. Several other targets have been identified, and include the E2 subunits of branched-chain 2-oxoacid dehydrogenase complex (BCOADC) and 2-oxoglutarate dehydrogenase complex (OGDC), and the E1α and E1β subunits of PDC [2, 10, 11, 23, 26–29]. One group in Newcastle has highlighted the significance of AMA, indicating that the majority of asymptomatic, non-cholestatic patients positive for AMA have histological features of PBC [22, 30].

PBC is generally a slowly progressive disease, but the disease course is unpredictable [1–3, 6–9]. The treatment of choice is with ursodeoxycholic acid (UDCA) administered at an appropriate dose (13–15 mg/kg/day) [1–3, 6–9]. UDCA administered in the early phase of the disease can dramatically slow the disease progression, and improve the quality of life in the majority of the patients [1–3, 6–9].

Several risk factors for the development of PBC have been identified in large epidemiological studies (Table 1) [31–35]. Risk factors which have been consistently noted include recurrent urinary tract infections (UTI), cigarette smoking, and estrogen deficiency. Female sex as well as being a first degree relative of a patient with PBC also increases the risk of disease development [31–34, 36]. Genetic and genome-wide association studies (GWAS) have identified several disease susceptibility genes, but it is likely that the development of PBC is multifactorial, with both genetic and environmental factors being involved [37]. Risk reduction, such as hormonal therapy or smoking cessation, and the aggressive treatment of recurrent UTI, has been suggested in PBC patients, based on known risk factors [31–34, 38]. There are currently no reliable prognostic indicators for PBC [2, 6–9].

Table 1 Evidence suggesting a role of xenobiotics in the development of primary biliary cirrhosis (PBC)

This review will examine two environmental and genetic factors which appear to play a role in the development of PBC, but may also explain the female predominance of the disease. These factors include xenobiotics, which are extrinsic chemical components found within the body, as well as monosomy X [39].

Genetics and monosomy X

Twin studies have demonstrated that there is a high concordance of PBC among monozygotic twins, and a low concordance among dizygotic twins, which is suggestive of a strong genetic influence [40]. Genetic studies as well as GWAS have identified several genes which appear to be related to PBC [41–49]. In a North American cohort, Hirschfield and colleagues [50] found a strong association between with HLA DQB1, as well as at the IL12A, IL12RB2, STAT4 and CTLA4 loci. That group also identified IRF5-TNPO3, 17q12-21, and MMEL1 loci to also be associated with PBC [51]. A study of Japanese patients found an association with 17q12-21, but no association was found with IL12A, IL12RB2 or IRF5-TNPO3 in that study [47]. Similar findings were reported by Liu and colleagues [52]. Other genes associated with PBC include STAT4, DENND1B, CD80, IL7R, CXCR5, TNFRSF1A, CLEC16A and NFKB1 [53]. More recent data provided strong evidence in support of genetic association of specific HLA and non-HLA genes with PBC [45].

As mentioned above, there is a higher risk for PBC development in female relatives of PBC patients, which further suggests that genetic factors are involved in the disease development, and that these factors may be related to the X chromosome [54–65]. As well, genes involved in immunological tolerance are located on the X chromosome, which may also partially explain the predominance of autoimmune disease in females in general [65–68]. Interestingly, a higher frequency of monosomy X in peripheral leukocytes has been found in patients with PBC [39, 66, 67, 69, 70]. One study examined the prevalence of X monosomy via fluorescence in situ hybridisation of peripheral white blood cells in 100 female PBC patients, and 50 patients with chronic hepatitis C and 50 healthy controls [66]. It was found that women with PBC had a higher frequency of monosomy X compared to pathological and healthy controls [66]. That study also noted that the frequency of monosomy X increased with age, which may also contribute to the explanation as to why PBC occurs predominantly in middle-aged women, and is virtually absent in the paediatric age [71–74]. Another study also noted that in addition to PBC, X monosomy was also a feature of systemic sclerosis (SSc) and autoimmune thyroid disease, which are often found to be concomitant in PBC patients [67]. As well, X monosomy was more frequent in peripheral T and B lymphocytes than other blood cell types [67]. Using similar techniques to those above, Svyryd et al. [75] determined the rate of X monosomy in Reynold’s syndrome, which is a laminopathy with combined PBC and progressive SSc. X monosomy was found in 12.1 % of Reynold’s syndrome, 10 % in PBC, 9.2 % in SSc, and 6.4 % in age-matched healthy controls [75]. Interestingly, an additive effect (89.1 %) of PBC and SSc on the prevalence of X monosomy in Reynold’s syndrome patients was noted [75]. As well, the prevalence of X monosomy did not appear dependent on the time of evolution of the autoimmune disease studied [75].

Miozzo et al. [76] set out to determine the mechanism of X chromosome inactivation in 166 female PBC patients and 266 age-matched controls, consisting of both healthy subjects and liver disease patients. That study found that in PBC, X chromosome loss occurred more frequently and in a preferential fashion [76]. That study also highlighted the need to determine the origin of the remaining X chromosome, as imprinting may also play a role in disease development [76]. A recent study by Mitchell et al. [77] tested whether susceptibility to PBC is modified by one or more X-linked gene with variable X chromosome inactivation status, given that 10 % of genes on the X chromosome escape X chromosome inactivation. Peripheral blood mRNA and DNA samples were obtained from monozygotic twin sets both discordant and concordant for PBC [77]. CLIC2 and PIN4 were identified as being consistently down-regulated in the affected twin of discordant pairs [77]. It is likely that more complex epigenetic factors play a role in the development of PBC.

Xenobiotics

Xenobiotics are exogenous chemical compounds that are found within an organism, which are not normally present or expected to be present within that organism. Examples of xenobiotics include antibiotics and pharmaceutical compounds, pollutants, and pesticides to name a few. Xenobiotics have been proposed as a possible environmental trigger of PBC in genetically susceptible individuals [78–83]. Indeed, clusters of PBC have been noted in regions of Newcastle and New York polluted with toxic waste (Table 1) [81]. It is believed that xenobiotics may induce a loss of tolerance by altering or forming complexes with self-peptides, thus eliciting immune response. In PBC, it is possible that a particular chemical compound may alter mitochondrial autoepitopes, or act as mimics to mitochondrial antigens, in a manner similar to molecular mimicry [84, 85]. Leung et al. [86] were capable of inducing AMAs in an animal model when immunised with 6-bromohexanoate conjugated with bovine albumin. Amano et al. [87] demonstrated that continued immunisation of mice with xenobiotics resulted in loss of tolerance to mitochondrial antigens, although this was reversible when exposure to the xenobiotic was stopped. The same group of researchers [88] suggest that the lipoyl domain of the immunodominant E2 component of PDC-E2 is replaced by a chemical xenobiotic mimic, which is capable of breaking self-tolerance in PBC. That group examined 107 potential xenobiotic mimics by coupling them to the lysine residues of the immunodominant region of PDC-E2, and spotted on microarray slides [88]. This was followed by a reactivity assay with sera from 47 PBC patients, 15 with primary sclerosing cholangitis, and 20 healthy volunteers [88]. Thirty-three of 107 xenobiotics had a significant IgG reactivity against PBC sera when compared with control sera, with nine of those compounds being more reactive than the native lipoylated peptide [88]. In addition, absorption studies demonstrated cross-reactivity with lipoic acid in eight of the nine compounds, including 2-octynoic acid, which is used in perfumes, lipsticks and food flavorings [88]. This is of interest given that several epidemiological studies have indicated hair-dye and nail polish use as being a risk factor for the development of PBC [31–34]. A recent study by Naiyanetr et al. [89] showed that AMA-positive PBC sera reacted against 6,8-bis(acetylthio)octanoic acid, which suggests that chemical modification of the lipoyl ring converts lipoic acid to its reduced form which promotes modification by xenobiotics. Therefore, common electrophilic agents such as acetaminophen (paracetamol) and non-steroidal anti-inflammatory drugs may induce xenobiotic modification in genetically susceptible individuals, followed by the production of AMA and possibly PBC [89]. This is of interest given that a recent study has indicated that patients who take an excessive amount of acetaminophen develop AMA with PBC-like specificity, and that metabolites of acetaminophen are immunoreactive with AMA [90]. The above studies demonstrate a possible correlation between risk factors of PBC and the molecular pathways which may be involved in a loss of immunological tolerance. The fact that several of the implicated xenobiotics are components of cosmetics raises the possible link between xenobiotic exposure and female predominance of PBC (Table 1).

Conclusions

Monsomy X and xenobiotics constitute the X-factor of PBC, which takes into account components of genetic susceptibility and environmental triggers, respectively. These components appear to relate to the female predominance of PBC. Further studies are needed to examine the role of epigenetic factors in the induction of PBC. As well, the identification of plausible xenobiotics is needed to allow for testing of these components in PBC patients. An understanding of the genetic components and environmental influences involved in the pathogenesis of PBC, may allow for proper identification of at-risk individuals, as well as possible prevention of the disease development with careful counselling (if possible) in regards to environmental exposure.

Abbreviations

AMA:

Antimitochondrial antibodies

ANA:

Antinuclear antibodies

PBC:

Primary biliary cirrhosis

PDC-E2:

Pyruvate dehydrogenase complex

SSc:

Systemic sclerosis

UTI:

Urinary tract infection

References

  1. Hohenester S, Oude-Elferink RP, Beuers U (2009) Primary biliary cirrhosis. Semin Immunopathol 31:283–307

    Article  PubMed  PubMed Central  Google Scholar 

  2. Kaplan MM, Gershwin ME (2005) Primary biliary cirrhosis. N Engl J Med 353:1261–1273

    Article  PubMed  CAS  Google Scholar 

  3. Neuberger J (1997) Primary biliary cirrhosis. Lancet 350:875–879

    Article  PubMed  CAS  Google Scholar 

  4. Invernizzi P, Mackay IR (2008) Autoimmune liver diseases. World J Gastroenterol 14:3290–3291

    Article  PubMed  PubMed Central  Google Scholar 

  5. Invernizzi P, Selmi C, Gershwin ME (2010) Update on primary biliary cirrhosis. Dig Liver Dis 42:401–408

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  6. Hemminki K, Li X, Sundquist K et al (2009) Shared familial aggregation of susceptibility to autoimmune diseases. Arthritis Rheum 60:2845–2847

    Article  PubMed  Google Scholar 

  7. Kumagi T, Heathcote EJ (2008) Primary biliary cirrhosis. Orphanet J Rare Dis 3:1

    Article  PubMed  PubMed Central  Google Scholar 

  8. Lindor KD, Gershwin ME, Poupon R et al (2009) Primary biliary cirrhosis. Hepatology 50:291–308

    Article  PubMed  Google Scholar 

  9. Poupon R (2010) Primary biliary cirrhosis. J Hepatol 52:745–758

    Article  PubMed  Google Scholar 

  10. Bogdanos DP, Baum H, Vergani D (2003) Antimitochondrial and other autoantibodies. Clin Liver Dis 7:759–777

    Article  PubMed  Google Scholar 

  11. Bogdanos DP, Invernizzi P, Mackay IR et al (2008) Autoimmune liver serology: current diagnostic and clinical challenges. World J Gastroenterol 14:3374–3387

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  12. Mackay IR (1958) Primary biliary cirrhosis showing a high titer of autoantibody: report of a case. N Engl J Med 258:185–188

    Article  PubMed  CAS  Google Scholar 

  13. Bogdanos DP, Komorowski L (2011) Disease-specific autoantibodies in primary biliary cirrhosis. Clin Chim Acta 412:502–512

    Article  PubMed  CAS  Google Scholar 

  14. Invernizzi P, Selmi C, Ranftler C et al (2005) Antinuclear antibodies in primary biliary cirrhosis. Semin Liver Dis 25:298–310

    Article  PubMed  Google Scholar 

  15. Rigopoulou EI, Bogdanos DP, Liaskos C et al (2007) Anti-mitochondrial antibody immunofluorescent titres correlate with the number and intensity of immunoblot-detected mitochondrial bands in patients with primary biliary cirrhosis. Clin Chim Acta 380:118–121

    Article  PubMed  CAS  Google Scholar 

  16. Vergani D, Bogdanos DP (2003) Positive markers in AMA-negative PBC. Am J Gastroenterol 98:241–243

    Article  PubMed  Google Scholar 

  17. Walker JG, Doniach D, Roitt IM et al (1965) Serological tests in diagnosis of primary biliary cirrhosis. Lancet 1:827–831

    Article  PubMed  CAS  Google Scholar 

  18. Kisand KE, Metskula K, Kisand KV et al (2001) The follow-up of asymptomatic persons with antibodies to pyruvate dehydrogenase in adult population samples. J Gastroenterol 36:248–254

    Article  PubMed  CAS  Google Scholar 

  19. Mattalia A, Quaranta S, Leung PS et al (1998) Characterization of antimitochondrial antibodies in health adults. Hepatology 27:656–661

    Article  PubMed  CAS  Google Scholar 

  20. Tunbridge WM, Evered DC, Hall R et al (1977) The spectrum of thyroid disease in a community: the Whickham survey. Clin Endocrinol 7:481–493

    Article  CAS  Google Scholar 

  21. Dahnrich C, Pares A, Caballeria L et al (2009) New ELISA for detecting primary biliary cirrhosis-specific antimitochondrial antibodies. Clin Chem 55:978–985

    Article  PubMed  Google Scholar 

  22. Mitchison HC, Bassendine MF, Hendrick A et al (1986) Positive antimitochondrial antibody but normal alkaline phosphatase: is this primary biliary cirrhosis? Hepatology 6:1279–1284

    Article  PubMed  CAS  Google Scholar 

  23. Leung PS, Coppel RL, Ansari A et al (1997) Antimitochondrial antibodies in primary biliary cirrhosis. Semin Liver Dis 17:61–69

    Article  PubMed  CAS  Google Scholar 

  24. Liu H, Norman GL, Shums Z et al (2010) PBC screen: an IgG/IgA dual isotype ELISA detecting multiple mitochondrial and nuclear autoantibodies specific for primary biliary cirrhosis. J Autoimmun 35:436–442

    Article  PubMed  CAS  Google Scholar 

  25. Rowley MJ, McNeilage LJ, Armstrong JM et al (1991) Inhibitory autoantibody to a conformational epitope of the pyruvate dehydrogenase complex, the major autoantigen in primary biliary cirrhosis. Clin Immunol Immunopathol 60:356–370

    Article  PubMed  CAS  Google Scholar 

  26. Van de Water J, Fregeau D, Davis P et al (1988) Autoantibodies of primary biliary cirrhosis recognize dihydrolipoamide acetyltransferase and inhibit enzyme function. J Immunol 141:2321–2324

    PubMed  Google Scholar 

  27. Dubel L, Tanaka A, Leung PS et al (1999) Autoepitope mapping and reactivity of autoantibodies to the dihydrolipoamide dehydrogenase-binding protein (E3BP) and the glycine cleavage proteins in primary biliary cirrhosis. Hepatology 29:1013–1018

    Article  PubMed  CAS  Google Scholar 

  28. Palmer JM, Jones DE, Quinn J et al (1999) Characterization of the autoantibody responses to recombinant E3 binding protein (protein X) of pyruvate dehydrogenase in primary biliary cirrhosis. Hepatology 30:21–26

    Article  PubMed  CAS  Google Scholar 

  29. Bogdanos DP, Vergani D (2006) Origin of cross-reactive autoimmunity in primary biliary cirrhosis. Liver Int 26:633–635

    Article  PubMed  CAS  Google Scholar 

  30. Metcalf JV, Mitchison HC, Palmer JM et al (1996) Natural history of early primary biliary cirrhosis. Lancet 348:1399–1402

    Article  PubMed  CAS  Google Scholar 

  31. Corpechot C, Chretien Y, Chazouilleres O et al (2010) Demographic, lifestyle, medical and familial factors associated with primary biliary cirrhosis. J Hepatol 53:162–169

    Article  PubMed  Google Scholar 

  32. Gershwin ME, Selmi C, Worman HJ et al (2005) Risk factors and comorbidities in primary biliary cirrhosis: a controlled interview-based study of 1032 patients. Hepatology 42:1194–1202

    Article  PubMed  PubMed Central  Google Scholar 

  33. Parikh-Patel A, Gold EB, Worman H et al (2001) Risk factors for primary biliary cirrhosis in a cohort of patients from the United States. Hepatology 33:16–21

    Article  PubMed  CAS  Google Scholar 

  34. Prince MI, Ducker SJ, James OF (2010) Case-control studies of risk factors for primary biliary cirrhosis in two United Kingdom populations. Gut 59:508–512

    Article  PubMed  CAS  Google Scholar 

  35. Smyk D, Rigopoulou EI, Bizzaro N et al (2012) Hair dyes as a risk for autoimmunity: from systemic lupus erythematosus to primary biliary cirrhosis. Autommun Highlights

  36. Smyk D, Grammatikopoulos T, Daponte A et al (2011) Fetomaternal alloimmunity as a cause of liver disease. Autoimmun Highlights 2:21–28

    Article  CAS  Google Scholar 

  37. Disanto G, Chaplin G, Morahan JM et al (2012) Month of birth, vitamin D and risk of immune mediated disease: a case control study. BMC Med 10:69

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  38. Bogdanos DP, Smyk DS, Rigopoulou EI et al (2012) Smoking as a risk factor for autoimmune liver disease: what we can learn from primary biliary cirrhosis. Ann Hepatol Off J Mexican Assoc Hepatol 11:7–14

    Google Scholar 

  39. Selmi C, Invernizzi P, Miozzo M et al (2004) Primary biliary cirrhosis: does X mark the spot? Autoimmun Rev 3:493–499

    Article  PubMed  CAS  Google Scholar 

  40. Selmi C, Mayo MJ, Bach N et al (2004) Primary biliary cirrhosis in monozygotic and dizygotic twins: genetics, epigenetics, and environment. Gastroenterology 127:485–492

    Article  PubMed  Google Scholar 

  41. Hirschfield GM, Invernizzi P (2011) Progress in the genetics of primary biliary cirrhosis. Semin Liver Dis 31:147–156

    Article  PubMed  CAS  Google Scholar 

  42. Invernizzi P (2009) Future directions in genetic for autoimmune diseases. J Autoimmun 33:1–2

    Article  PubMed  CAS  Google Scholar 

  43. Invernizzi P, Gershwin ME (2009) The genetics of human autoimmune disease. J Autoimmun 33:290–299

    Article  PubMed  CAS  Google Scholar 

  44. Invernizzi P, Ransom M, Raychaudhuri S et al (2012) Classical HLA-DRB1 and DPB1 alleles account for HLA associations with primary biliary cirrhosis. Genes Immun 13:461–468

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  45. Juran BD, Hirschfield GM, Invernizzi P et al. (2012) 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

  46. Invernizzi P, Selmi C, Poli F et al (2008) Human leukocyte antigen polymorphisms in Italian primary biliary cirrhosis: a multicenter study of 664 patients and 1992 healthy controls. Hepatology 48:1906–1912

    Article  PubMed  PubMed Central  Google Scholar 

  47. Tanaka A, Invernizzi P, Ohira H et al (2011) Replicated association of 17q12-21 with susceptibility of primary biliary cirrhosis in a Japanese cohort. Tissue Antigens 78:65–68

    Article  PubMed  CAS  Google Scholar 

  48. Smyk D, Mytilinaiou MG, Milkiewicz P et al (2012) Towards systemic sclerosis and away from primary biliary cirrhosis: the case of PTPN22. Autommun Highlights 3:1–9

    Article  CAS  Google Scholar 

  49. Kronenberger B, Rudloff I, Bachmann M et al (2012) Interleukin-22 predicts severity and death in advanced liver cirrhosis: a prospective cohort study. BMC Med 10:102

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  50. Hirschfield GM, Liu X, Xu C et al (2009) Primary biliary cirrhosis associated with HLA, IL12A, and IL12RB2 variants. N Engl J Med 360:2544–2555

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  51. Hirschfield GM, Liu X, Han Y et al (2010) Variants at IRF5-TNPO3, 17q12-21 and MMEL1 are associated with primary biliary cirrhosis. Nat Genet 42:655–657

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  52. Liu X, Invernizzi P, Lu Y et al (2010) Genome-wide meta-analyses identify three loci associated with primary biliary cirrhosis. Nat Genet 42:658–660

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  53. Mells GF, Floyd JA, Morley KI et al (2011) Genome-wide association study identifies 12 new susceptibility loci for primary biliary cirrhosis. Nat Genet 43:329–332

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  54. Bach N, Schaffner F (1994) Familial primary biliary cirrhosis. J Hepatol 20:698–701

    Article  PubMed  CAS  Google Scholar 

  55. Brind AM, Bray GP, Portmann BC et al (1995) Prevalence and pattern of familial disease in primary biliary cirrhosis. Gut 36:615–617

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  56. Fagan E, Williams R, Cox S (1977) Primary biliary cirrhosis in mother and daughter. Br Med J 2:1195

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  57. Floreani A, Naccarato R, Chiaramonte M (1997) Prevalence of familial disease in primary biliary cirrhosis in Italy. J Hepatol 26:737–738

    Article  PubMed  CAS  Google Scholar 

  58. Jaup BH, Zettergren LS (1980) Familial occurrence of primary biliary cirrhosis associated with hypergammaglobulinemia in descendants: a family study. Gastroenterology 78:549–555

    PubMed  CAS  Google Scholar 

  59. Jones DE, Watt FE, Metcalf JV et al (1999) Familial primary biliary cirrhosis reassessed: a geographically-based population study. J Hepatol 30:402–407

    Article  PubMed  CAS  Google Scholar 

  60. Lazaridis KN, Juran BD, Boe GM et al (2007) Increased prevalence of antimitochondrial antibodies in first-degree relatives of patients with primary biliary cirrhosis. Hepatology 46:785–792

    Article  PubMed  Google Scholar 

  61. Tsuji K, Watanabe Y, Van de Water J et al (1999) Familial primary biliary cirrhosis in Hiroshima. J Autoimmun 13:171–178

    Article  PubMed  CAS  Google Scholar 

  62. Invernizzi P, Selmi C, Mackay IR et al (2005) From bases to basis: linking genetics to causation in primary biliary cirrhosis. Clin Gastroenterol Hepatol 3:401–410

    Article  PubMed  CAS  Google Scholar 

  63. Invernizzi P (2007) Role of X chromosome defects in primary biliary cirrhosis. Hepatol Res 37(Suppl 3):S384–S388

    Article  PubMed  Google Scholar 

  64. Milkiewicz P, Heathcote J (2005) Primary biliary cirrhosis in a patient with Turner syndrome. Can J Gastroenterol 19:631–633

    PubMed  Google Scholar 

  65. Invernizzi P, Pasini S, Selmi C et al (2009) Female predominance and X chromosome defects in autoimmune diseases. J Autoimmun 33:12–16

    Article  PubMed  CAS  Google Scholar 

  66. Invernizzi P, Miozzo M, Battezzati PM et al (2004) Frequency of monosomy X in women with primary biliary cirrhosis. Lancet 363:533–535

    Article  PubMed  Google Scholar 

  67. Invernizzi P, Miozzo M, Selmi C et al (2005) X chromosome monosomy: a common mechanism for autoimmune diseases. J Immunol 175:575–578

    Article  PubMed  CAS  Google Scholar 

  68. Invernizzi P, Miozzo M, Oertelt-Prigione S et al (2007) X monosomy in female systemic lupus erythematosus. Ann N Y Acad Sci 1110:84–91

    Article  PubMed  CAS  Google Scholar 

  69. Selmi C, Invernizzi P, Zuin M et al (2005) Genes and (auto)immunity in primary biliary cirrhosis. Genes Immun 6:543–556

    Article  PubMed  CAS  Google Scholar 

  70. Bogdanos DP, Smyk DS, Rigopoulou EI et al (2012) Twin studies in autoimmune disease: genetics, gender and environment. J Autoimmun 38:J156–J169

    Article  PubMed  Google Scholar 

  71. Invernizzi P, Alessio MG, Smyk DS et al (2012) Autoimmune hepatitis type 2 associated with an unexpected and transient presence of primary biliary cirrhosis-specific antimitochondrial antibodies: a case study and review of the literature. BMC Gastroenterol 12:92

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  72. Smyk DS, Rigopoulou EI, Lleo A et al (2011) Immunopathogenesis of primary biliary cirrhosis: an old wives’ tale. Immun Ageing I A 8:12

    Article  PubMed  CAS  Google Scholar 

  73. Smyk DS, Rigopoulou EI, Pares A et al (2012) Sex differences associated with primary biliary cirrhosis. Clin Dev Immunol 2012:610504

    Article  PubMed  PubMed Central  Google Scholar 

  74. Smyk DS, Rigopoulou EI, Pares A et al (2012) Familial primary biliary cirrhosis: like mother, like daughter? Acta Gastroenterol Belg 75:203–209

    PubMed  CAS  Google Scholar 

  75. Svyryd Y, Hernandez-Molina G, Vargas F et al (2012) X chromosome monosomy in primary and overlapping autoimmune diseases. Autoimmun Rev 11:301–304

    Article  PubMed  CAS  Google Scholar 

  76. Miozzo M, Selmi C, Gentilin B et al (2007) Preferential X chromosome loss but random inactivation characterize primary biliary cirrhosis. Hepatology 46:456–462

    Article  PubMed  CAS  Google Scholar 

  77. Mitchell MM, Lleo A, Zammataro L et al (2011) Epigenetic investigation of variably X chromosome inactivated genes in monozygotic female twins discordant for primary biliary cirrhosis. Epigenetics 6:95–102

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  78. Gershwin ME, Mackay IR (2008) The causes of primary biliary cirrhosis: convenient and inconvenient truths. Hepatology 47:737–745

    Article  PubMed  Google Scholar 

  79. Selmi C, Gershwin ME (2009) The role of environmental factors in primary biliary cirrhosis. Trends Immunol 30:415–420

    Article  PubMed  CAS  Google Scholar 

  80. Leung PS, Iwayama T, Coppel RL et al (1990) Site-directed mutagenesis of lysine within the immunodominant autoepitope of PDC-E2. Hepatology 12:1321–1328

    Article  PubMed  CAS  Google Scholar 

  81. Smyk D, Mytilinaiou MG, Rigopoulou EI et al (2010) PBC triggers in water reservoirs, coal mining areas and waste disposal sites: from Newcastle to New York. Dis Markers 29:337–344

    Article  PubMed  PubMed Central  Google Scholar 

  82. Smyk D, Rigopoulou EI, Baum H et al (2012) Autoimmunity and environment: am I at risk? Clin Rev Allergy Immunol 42(2):199–212

    Article  PubMed  CAS  Google Scholar 

  83. Smyk DS, Rigopoulou EI, Muratori L et al (2012) Smoking as a risk factor for autoimmune liver disease: what we can learn from primary biliary cirrhosis. Ann Hepatol 11:7–14

    PubMed  CAS  Google Scholar 

  84. Bogdanos DP, Mieli-Vergani G, Vergani D (2000) Virus, liver and autoimmunity. Dig Liver Dis 32:440–446

    Article  PubMed  CAS  Google Scholar 

  85. Oldstone MB (1987) Molecular mimicry and autoimmune disease. Cell 50:819–820

    Article  PubMed  CAS  Google Scholar 

  86. Leung PS, Quan C, Park O et al (2003) Immunization with a xenobiotic 6-bromohexanoate bovine serum albumin conjugate induces antimitochondrial antibodies. J Immunol 170:5326–5332

    Article  PubMed  CAS  Google Scholar 

  87. Amano K, Leung PS, Xu Q et al (2004) Xenobiotic-induced loss of tolerance in rabbits to the mitochondrial autoantigen of primary biliary cirrhosis is reversible. J Immunol 172:6444–6452

    Article  PubMed  CAS  Google Scholar 

  88. Amano K, Leung PS, Rieger R et al (2005) Chemical xenobiotics and mitochondrial autoantigens in primary biliary cirrhosis: identification of antibodies against a common environmental, cosmetic, and food additive, 2-octynoic acid. J Immunol 174:5874–5883

    Article  PubMed  CAS  Google Scholar 

  89. Naiyanetr P, Butler JD, Meng L et al (2011) Electrophile-modified lipoic derivatives of PDC-E2 elicits anti-mitochondrial antibody reactivity. J Autoimmun 37:209–216

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  90. Leung PS, Lam K, Kurth MJ et al (2012) Xenobiotics and autoimmunity: does acetaminophen cause primary biliary cirrhosis? Trends Mol Med 18(10):577–582

    Article  PubMed  CAS  PubMed Central  Google Scholar 

Download references

Conflict of interest

None.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Pietro Invernizzi.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Bianchi, I., Lleo, A., Bernuzzi, F. et al. The X-factor in primary biliary cirrhosis: monosomy X and xenobiotics. Autoimmun Highlights 3, 127–132 (2012). https://doi.org/10.1007/s13317-012-0043-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13317-012-0043-2

Keywords