ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Pathogenesis of primary biliary cholangitis (primary biliary cirrhosis)

Pathogenesis of primary biliary cholangitis (primary biliary cirrhosis)
Literature review current through: Jan 2024.
This topic last updated: Nov 10, 2023.

INTRODUCTION — Primary biliary cholangitis (PBC; previously referred to as primary biliary cirrhosis) is characterized by a T-lymphocyte-mediated attack on small intralobular bile ducts (picture 1A-B). A continuous assault on the bile duct epithelial cells leads to their gradual destruction and eventual disappearance (picture 2). The sustained loss of intralobular bile ducts causes the signs and symptoms of cholestasis, and eventually results in cirrhosis and liver failure [1-3]. (See "Clinical manifestations, diagnosis, and prognosis of primary biliary cholangitis".)

This topic will review the pathogenesis of PBC. The diagnosis and management of PBC are discussed separately. (See "Clinical manifestations, diagnosis, and prognosis of primary biliary cholangitis" and "Overview of the management of primary biliary cholangitis".)

CLUES ABOUT ETIOLOGY BASED ON THE EPIDEMIOLOGY OF PBC — The precise cause of PBC is unknown but, as with other autoimmune diseases, is related to genetic susceptibility and environmental factors [4-6]. A number of environmental causes have been implicated, including several bacteria, viruses, toxins, and drugs, which are described below [7-11]. Some of the most compelling evidence for an environmental factor has been derived from epidemiologic studies, which have demonstrated geographic clustering, clustering of cases across time, and seasonal variation in the diagnosis of PBC [9,10].

As examples, the incidence of PBC has been estimated to be 200 to 251 per million in South Wales and Northeast England, less than 25 per million in Canada and Australia, and almost zero in Sub-Saharan Africa and India [12-16]. A limitation of these studies is that the varying incidence rates may in part be due to genetic differences in the populations and to differences in study methodology. However, that an environmental factor may be involved was strongly implicated in a study from a well-defined geographical population cohort in Northern England [17] and around toxic waste sites in New York City [8]. Analysis of cases demonstrated unequivocal clustering in well-demarcated geographic regions.

The unanswered question in PBC is what causes or triggers the T-cell attack on small bile duct epithelial cells? Available data suggest that PBC is an autoimmune disease [18,19]. Like other better characterized autoimmune diseases, there appear to be at least two distinct requirements for PBC to develop: genetic susceptibility; and a triggering event that initiates the autoimmune attack on bile duct cells.

The prevalence of PBC in families with one affected member is estimated to be 100-fold greater than that in the general population [20,21]. However, the disorder is not inherited in any simple recessive or dominant pattern. Familial occurrences of the disease have included sisters [22], brothers [23], brothers and sisters [24], and parent and child [25]. In addition, unaffected family members are more likely than controls to have impaired T-cell regulation [26] and increased numbers of circulating autoantibodies [23]. However, there is not a significant increase in antimitochondrial antibody, the serologic marker of PBC, in healthy family members if purified recombinant mitochondrial autoantigens are used in the assay [27]. (See 'Antimitochondrial antibodies' below.)

Genetic susceptibility — There have been extensive genome-wide association studies (GWAS) in patients with PBC, and a number of associations have been documented [28]. However, while such associations are of considerable interest, they have not yet been translated into useful clinical testing [29-31]. There is a weak association between PBC and haplotype HLA-DR8 [32-34] and the HLA-DPB1 gene in some populations [35,36]. Accumulated data suggest that there may be an inherited abnormality of immune regulation, perhaps an inability to suppress an inflammatory attack on small bile ducts once it is initiated [32,35,37]. One study found an association of PBC with a variant in the CTLA4 gene, which encodes a co-inhibitory immunoreceptor involved in self-tolerance [38]. Another report found a significant association with genetic variants at the HLA class II, IL12A, and IL12RB2 loci (which have a role in interleukin-12 immunoregulatory signaling) [31]. The importance of IL12 has been underscored in animal models and in a case report of a child with congenital IL12 deficiency who developed PBC [39]. Other susceptibility loci that have been identified include SPIB (which encodes for a transcription factor involved in B-cell receptor signaling and T-cell lineage decisions), IRF5 (which encodes for interferon regulatory factor 5), TNPO3 (which encodes for transportin 3), and MMEL1 (which encodes for a metallo-endopeptidase) [40,41]. Interestingly, IRF5 and TNPO3 have also been implicated in the pathogenesis of systemic lupus erythematosus, systemic sclerosis and Sjogren syndrome, suggesting that there is a genetic overlap in PBC and these disorders. The concordance of PBC in identical twins is approximately 50 percent, somewhat higher than the concordance of autoimmunity in identical twins with lupus and rheumatoid arthritis [42,43].

Why the disease primarily affects females is unclear. A plausible theory relates to immunological tolerance, a component of which depends upon genes located on the X chromosome [44]. One study suggested that females with PBC were more likely to have monosomy of the X chromosome (a marker reflecting haploinsufficiency for specific X-linked genes) compared with healthy controls or those with hepatitis C [45].

Finally, there have been suggestions for an epigenetic role in the pathogenesis of PBC, (ie, methylation), and therefore modification of DNA [46].

MOLECULAR MIMICRY — Molecular mimicry refers to a phenomenon where antigens evoking an immune response have enough similarity to endogenous proteins to incite an autoimmune reaction. Such a mechanism has been widely proposed to initiate autoimmunity in patients with primary biliary cholangitis (PBC). Several candidate inciting agents (including bacteria, viruses and various chemicals) have been proposed, but none has been proven definitively.

Infections — An infectious etiology for triggering PBC has been suspected, although a specific etiologic agent has not been unequivocally identified. Several agents have been implicated; some have been disproven while others remain topics of investigation.

It is possible that infection and the presence of antimitochondrial antibodies (AMA) (see 'Antimitochondrial antibodies' below) may be pathogenetically linked. A study comparing peripheral blood mononuclear cells from patients with PBC with controls found that patients with PBC had a much higher number of inducible IgM-producing B-cells and that each B-cell produced a greater quantity of IgM protein compared with controls [47]. Induction of the B-cells was achieved by stimulation with unmethylated CpG DNA motifs (non-proteinaceous repeated sequences found frequently in bacteria). The authors hypothesized that patients with PBC are hyperresponsive with respect to IgM production and that the heightened IgM production may reflect how various microorganisms interact with the immune system in patients with PBC. Interestingly, there is a significant increase in AMA-specific plasmablasts in the blood of patients with PBC [48].

Retroviruses — Data from one laboratory suggested that infection with a retrovirus may be associated with PBC in some patients. However, these data have not been replicated [49] and have largely been disregarded.

Biochemical improvement from combination antiretroviral treatment was observed in an older open-label pilot study (also from the same group), a finding that has never been confirmed [50].

Propionibacterium acnes — A possible etiologic role for Propionibacterium acnes (P. acnes) was suggested in a study in which sequences of the organism were detected by PCR-amplifying granulomas from patients with PBC [51]. The significance of this observation remains uncertain but may relate to the appearance of granulomas in some patients with PBC.

Chlamydia pneumoniae — A pilot study reported that antigens to Chlamydia pneumoniae were detectable more often in the livers of patients with PBC compared with controls with other causes of liver disease, suggesting a possible etiologic role [52]. However, these findings were disputed in subsequent report using more detailed immunohistochemical analysis [53]. Furthermore, no benefit from treatment with tetracycline was observed in a pilot open-label study [54].

Escherichia coli — Antibodies reacting against the mitochondrial human pyruvate dehydrogenase complex crossreact with the E. coli pyruvate dehydrogenase complex implicating E. coli infection (particularly urinary tract infection) in the pathogenesis of PBC [55-59]. A problem with this theory is that antibodies against the E. coli are often in lower titers than against the human complex, and the E. coli antibodies are more frequent in patients with advanced disease (rather than being present in patients with early-stage disease, which would be more supportive of a role as an inciting agent). However, subsequent studies using different recombinant constructs of E. coli have suggested that infection with E. coli may be an early inciting event [60].

N. aromaticivorans — Novosphingobium aromaticivorans (N. aromaticivorans) is a ubiquitous gram-negative organism that metabolizes organic compounds and estrogens. Two of its proteins share homology with the E2 component of pyruvate dehydrogenase (PDC-E2), the major epitope recognized by antimitochondrial antibodies. A role for the bacteria in the pathogenesis of PBC was suggested in a pilot study in which 100 percent of 77 PBC patients (who were anti-PDC-E2 positive) had high antibody titers against the organism compared to none of 195 controls [61]. Reactivity to the organism was also found in another study [62]. These observations provide increasing evidence that exposure to N. aromaticivorans is associated with the development of PBC, the clinical implications of which are uncertain [63].

Lactobacillus — A case report described a patient who developed PBC after receiving intramuscular vaccination with a vaccine containing several lactobacillus species to treat recurrent vaginosis [64]. The patient had AMA-M2 antimitochondrial antibodies that reacted against the major PBC-specific mitochondrial auto-epitope (ie, pyruvate dehydrogenase complex E2) but reacted even more strongly against Lactobacillus delbrueckii. The authors speculated that the lactobacillus vaccination caused the development of PBC through molecular mimicry.

CHEMICAL EXPOSURE — Certain compounds (particularly halogenated hydrocarbons) are capable of inducing antibodies that have an affinity for the human pyruvate dehydrogenase complex (sometimes even having greater affinity for native mitochondrial antigens than for the halogenated hydrocarbon) [65,66]. In a rabbit model, one compound (bromohexanoate ester) induced high titers of antimitochondrial antibodies that were similar to those seen in humans with PBC [67]. However, the animals did not develop liver lesions similar to PBC. Such lesions did develop 18 months after immunization in a guinea pig model [68]. Additional data have led to the induction of histologic lesions that resemble PBC in mice immunized with a common component of cosmetics, 2-octynoic acid [69]. Residence near a toxic waste site was associated with PBC in one epidemiologic study [8].

ANTIMITOCHONDRIAL ANTIBODIES — Antimitochondrial antibodies are the serologic hallmark of PBC (see "Clinical manifestations, diagnosis, and prognosis of primary biliary cholangitis").

There are four principal autoantigens that are targets for antimitochondrial antibodies (AMA), which have collectively been referred to as the "M2" subtype of mitochondrial autoantigens: the E2 subunits of the pyruvate dehydrogenase complex, the branched chain 2-oxo-acid dehydrogenase complex, the ketoglutaric acid dehydrogenase complex, and the dihydrolipoamide dehydrogenase-binding protein [3]. Each of these autoantigens participates in oxidative phosphorylation (a process occurring in the inner mitochondrial membrane by which ATP is formed) and have a great deal of homology.

A major advance in our understanding of PBC occurred with the identification and cloning of these antigens [70]. Most AMA react against the dihydrolipoamide acetyltransferase component (E2 subunit) of pyruvate dehydrogenase (PDC-E2) [71-74]. PBC is the only disease in which there are B- and T-cells that are autoreactive against PDC-E2 (image 1). Thus far, all of the mitochondrial autoantigens screened have been targets of only the M2 antimitochondrial antibodies. Other antimitochondrial antibodies previously described, anti-M4, anti-M8, and anti-M9, are most likely artifacts of the methods used to detect them [75,76]. Human antimitochondrial antibodies inhibit the enzymatic activity of their target enzyme complexes in vitro [72].

Antimitochondrial antibodies are found in 95 percent of patients with PBC when using the most sensitive detection techniques [77], and have a specificity of 98 percent for the disease [71]. However, their role in pathogenesis is unclear as illustrated by the following observations:

AMA titers vary greatly (more than 20-fold) among patients but tend to be stable over time in an individual patient; antibody titers do not correlate with disease severity or rate of progression [78,79].

There is no apparent difference in the clinical spectrum or course of patients with PBC who are AMA-positive or AMA-negative [80].

Antimitochondrial antibodies raised in animals immunized with recombinant human pyruvate dehydrogenase do not cause bile duct damage or any recognizable disease [81].

Patients with either AMA-negative or AMA-positive PBC have a similar response to treatment with ursodeoxycholic acid or liver transplantation [82].

T-CELL MITOCHONDRIAL RESPONSE — Although the role of antimitochondrial antibodies in the pathogenesis of primary biliary cholangitis (PBC) is uncertain, accumulating data suggest a direct role for T-lymphocytes. CD4+ and CD8+ cells are present in high concentration in the portal triads of patients with PBC, often surrounding and infiltrating necrotic bile ducts [83-87]. Autoreactive T-lymphocytes from PBC patients are specific for PDC-E2 [88-91]. There is a 100- to 150-fold increase in precursor frequencies of PDC-E2-specific T-cells isolated from the liver and hilar lymph nodes compared to peripheral blood mononuclear cells in patients with PBC [84]. PDC-E2, normally found in the inner mitochondrial membrane of all cells, is aberrantly expressed in the luminal surface of bile duct epithelial cells only in patients with PBC [85].

The immunodominant epitope recognized by MHC class II restricted CD4+ T-cells in patients with PBC has been identified as amino acids 163 to 176 of PDC-E2 [90,91]. The immunodominant epitope recognized by MHC Class I restricted CD8+ T-cells has also been identified and is similar (amino acids 159 to 167 of PDC-E2) [87]. This peptide induces proliferation of specific MHC-class I restricted CD8+ cytotoxic T-lymphocytes from most HLA-A*0201 patients with PBC [92] and the frequency of cytotoxic T-lymphocytes recognizing this epitope is increased 10-fold in liver compared to blood in patients with PBC [87]. An alanine substitution at position 5 of this epitope significantly reduced peptide-specific cytotoxicity and interferon gamma production by cytotoxic CD8+ T-lymphocytes derived from peripheral blood monocytes of patients with PBC [93].

These data suggest that bile duct epithelial cells expressing this epitope are targeted by CD8+ T-lymphocytes in patients with PBC. Why bile duct epithelial cells express this epitope is still unknown. One possibility is that IgA is complexed to PDC-E2 in the serum of patients with PBC and this antigen-antibody complex is then transcytosed from the basal to apical side of the bile duct epithelial cell en route to its secretion in bile [85]. Serum IgA has been found at mitochondrial surfaces and immune complexes containing PDC-E2, and IgA have been found in bile duct lumens in patients with PBC [85,94].

In addition to the T-lymphocyte mediated destruction of small bile ducts, secondary damage to hepatocytes may occur from the accumulation in the liver of increased concentrations of potentially toxic substances, such as bile acids, which are normally secreted into bile (algorithm 1). The naturally occurring bile acids (cholic acid, chenodeoxycholic acid, and deoxycholic acid) are all detergents and can dissolve cell membranes if present in a sufficiently high concentration [95-97]; such toxic concentrations are often reached in cholestasis (figure 1) [98].

WHY TISSUE INJURY IN PBC IS CONFINED TO THE LIVER — A paradox of PBC is that the target mitochondrial antigens are present in every cell in the body while the immune attack appears to be highly specific for biliary epithelium. In addition, PBC may reoccur following liver transplantation. Since the donor liver contains donor MHC genes, this observation suggests that immune mechanisms, other than adaptive immunity, may perpetuate disease. Proposed mechanisms include innate immune responses [99]. One theory is that there may be qualitative differences in the processing of apoptotic bile-duct cells that account for the specificity, but the precise mechanisms involved are incompletely understood [100-102].

Another hypothesis proposes that any factor (whether it be an infection or a chemical) that damages bile duct epithelial cells may unmask an antigen in these cells that shares some epitopic similarities with pyruvate dehydrogenase complex E2 [103,104].

A molecule that shares some antigenic determinants with the E2 subunit of pyruvate dehydrogenase but is distinct from it has been identified on the luminal surface of biliary epithelial cells of patients with PBC early in the course of disease (image 1) [105,106]. The presence of this antigen, found only in bile duct epithelial cells of patients with PBC, could explain why cell injury in PBC is limited to biliary epithelial cells.

Expression of this autoantigen on the luminal surface of biliary epithelial cells may also provoke an antibody mediated attack by IgA antibodies, the antibody present in bile [105]. In addition, this E2-like antigen, together with the appropriate class II major histocompatibility complex antigens, and another molecule required for antigen presentation, BB1/B7, could be the target of activated CD8+ lymphocytes. All of these molecules are found in and around damaged bile ducts in PBC [106]. The E2 component appears in damaged bile ducts before the HLA class II antigens and BB1/B7 supporting a sequence whereby the bile duct epithelial cell injury precedes the T-cell mediated immune response. The histology of PBC is also consistent with T-lymphocyte mediated cytotoxicity as is the fact that the E2 antigens stimulate interleukin-2 production by cloned T-cells isolated from liver tissue [103].

ROLE OF RETAINED BILE ACIDS — The "foamy" degeneration of hepatocytes in primary biliary cholangitis (PBC) has been attributed to the noxious effects of bile acids (picture 3) [95]. Cholestasis per se causes increased expression of HLA class I antigens on hepatocytes, thereby rendering them better targets for activated T-lymphocytes. In this regard, there is increased evidence that the biliary system requires a bicarbonate umbrella, and that disruption of this umbrella makes biliary cells more prone to damage. In patients with PBC, emerging data suggest that the bicarbonate umbrella is defective. Whether the umbrella dysfunction is primary or secondary to disease is unknown [107].

Reversal of these pathogenetic events may explain the efficacy of ursodeoxycholic acid, a dihydroxy bile acid, which has been approved by the FDA for the treatment of PBC. Studies in both humans and experimental animals suggest that treatment with ursodeoxycholic acid reduces expression of HLA class I antigens by hepatocytes and also lessens the toxicity of retained naturally occurring bile acids such as cholic acid and chenodeoxycholic acid [96,97]. (See "Overview of the management of primary biliary cholangitis".)

FETAL MICROCHIMERISM — The persistence of allogeneic fetal cells in the maternal system has been termed fetal microchimerism. These cells have been postulated to have a role in the development of maternal diseases such as scleroderma [108], providing the rationale for studies of microchimerism in patients with PBC. Several studies on microchimerism in PBC have produced discordant results [109-112]. Thus, at the present time, the role of microchimerism is unproven [113].

CLINICAL PROGRESSION — The pathogenesis of biliary cirrhosis itself is straightforward after the immunologic injury and is similar to that which occurs in a variety of chronic cholestatic diseases, such as bile duct strictures and biliary atresia in children. Chronic impairment of bile flow leads to portal and parenchymal inflammation, liver cell necrosis, scarring, and eventually to cirrhosis and liver failure. The clinical progression, however, requires years, and at present there are no clear prognostic factors that predict the rate of liver failure in a given patient. Furthermore, it is likely that there are various immunological mechanisms at play during the progression of disease (ie, there may be different pathways involved in the inflammatory response, the destruction of bile ducts, and the development of fibrosis). (See "Clinical manifestations, diagnosis, and prognosis of primary biliary cholangitis".)

SUMMARY

Background – Primary biliary cholangitis is characterized by a T-lymphocyte-mediated attack on small intralobular bile ducts (picture 1A-B). A continuous assault on the bile duct epithelial cells leads to their gradual destruction and eventual disappearance (picture 2). (See 'Introduction' above.)

Pathogenesis – The pathogenesis of primary biliary cholangitis is incompletely understood but appears to involve genetic susceptibility and environmental factors. It is likely that more than one environmental trigger may elicit bile duct epithelial injury in a genetically susceptible individual. This will lead to both adaptive and innate immune responses that lead to portal inflammation and bile duct epithelial damage. (See 'Clues about etiology based on the epidemiology of PBC' above.)

Clinical progression – Once immune-mediated bile duct injury has been established, the disease progresses due to chronic cholestasis and secondary inflammation and scarring. (See 'Clinical progression' above.)

  1. Ludwig J. New concepts in biliary cirrhosis. Semin Liver Dis 1987; 7:293.
  2. Selmi C, Bowlus CL, Gershwin ME, Coppel RL. Primary biliary cirrhosis. Lancet 2011; 377:1600.
  3. Kaplan MM, Gershwin ME. Primary biliary cirrhosis. N Engl J Med 2005; 353:1261.
  4. Jones DE. Pathogenesis of primary biliary cirrhosis. J Hepatol 2003; 39:639.
  5. Gershwin ME, Selmi C, Worman HJ, et al. Risk factors and comorbidities in primary biliary cirrhosis: a controlled interview-based study of 1032 patients. Hepatology 2005; 42:1194.
  6. Naiyanetr P, Butler JD, Meng L, et al. Electrophile-modified lipoic derivatives of PDC-E2 elicits anti-mitochondrial antibody reactivity. J Autoimmun 2011; 37:209.
  7. Haydon GH, Neuberger J. PBC: an infectious disease? Gut 2000; 47:586.
  8. Ala A, Stanca CM, Bu-Ghanim M, et al. Increased prevalence of primary biliary cirrhosis near Superfund toxic waste sites. Hepatology 2006; 43:525.
  9. McNally RJ, Ducker S, James OF. Are transient environmental agents involved in the cause of primary biliary cirrhosis? Evidence from space-time clustering analysis. Hepatology 2009; 50:1169.
  10. McNally RJ, James PW, Ducker S, James OF. Seasonal variation in the patient diagnosis of primary biliary cirrhosis: further evidence for an environmental component to etiology. Hepatology 2011; 54:2099.
  11. Amano K, Leung PS, Rieger R, et al. 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 2005; 174:5874.
  12. Kingham JG, Parker DR. The association between primary biliary cirrhosis and coeliac disease: a study of relative prevalences. Gut 1998; 42:120.
  13. Witt-Sullivan H, Heathcote J, Cauch K, et al. The demography of primary biliary cirrhosis in Ontario, Canada. Hepatology 1990; 12:98.
  14. Watson RG, Angus PW, Dewar M, et al. Low prevalence of primary biliary cirrhosis in Victoria, Australia. Melbourne Liver Group. Gut 1995; 36:927.
  15. Kim WR, Lindor KD, Locke GR 3rd, et al. Epidemiology and natural history of primary biliary cirrhosis in a US community. Gastroenterology 2000; 119:1631.
  16. Podda M, Selmi C, Lleo A, et al. The limitations and hidden gems of the epidemiology of primary biliary cirrhosis. J Autoimmun 2013; 46:81.
  17. Prince MI, Chetwynd A, Diggle P, et al. The geographical distribution of primary biliary cirrhosis in a well-defined cohort. Hepatology 2001; 34:1083.
  18. Hirschfield GM, Gershwin ME. The immunobiology and pathophysiology of primary biliary cirrhosis. Annu Rev Pathol 2013; 8:303.
  19. Wang L, Wang FS, Chang C, Gershwin ME. Breach of tolerance: primary biliary cirrhosis. Semin Liver Dis 2014; 34:297.
  20. Bach N, Schaffner F. Familial primary biliary cirrhosis. J Hepatol 1994; 20:698.
  21. Tsuji K, Watanabe Y, Van De Water J, et al. Familial primary biliary cirrhosis in Hiroshima. J Autoimmun 1999; 13:171.
  22. James SP, Jones EA, Schafer DF, et al. Selective immunoglobulin A deficiency associated with primary biliary cirrhosis in a family with liver disease. Gastroenterology 1986; 90:283.
  23. Galbraith RM, Smith M, Mackenzie RM, et al. High prevalence of seroimmunologic abnormalities in relatives of patients with active chronic hepatitis or primary biliary cirrhosis. N Engl J Med 1974; 290:63.
  24. Schaffner F. Primary biliary cirrhosis. Clin Gastroenterol 1975; 4:351.
  25. Tong MJ, Nies KM, Reynolds TB, Quismorio FP. Immunological studies in familial primary biliary cirrhosis. Gastroenterology 1976; 71:305.
  26. Lan RY, Cheng C, Lian ZX, et al. Liver-targeted and peripheral blood alterations of regulatory T cells in primary biliary cirrhosis. Hepatology 2006; 43:729.
  27. Caldwell SH, Leung PS, Spivey JR, et al. Antimitochondrial antibodies in kindreds of patients with primary biliary cirrhosis: antimitochondrial antibodies are unique to clinical disease and are absent in asymptomatic family members. Hepatology 1992; 16:899.
  28. Gerussi A, Asselta R, Invernizzi P. Genetics of Primary Biliary Cholangitis. Clin Liver Dis 2022; 26:571.
  29. Juran BD, Hirschfield GM, Invernizzi P, 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.
  30. Kar SP, Seldin MF, Chen W, et al. Pathway-based analysis of primary biliary cirrhosis genome-wide association studies. Genes Immun 2013; 14:179.
  31. Gulamhusein AF, Lazaridis KN. Primary biliary cholangitis, DNA, and beyond: The Relative contribution of genes. Hepatology 2018; 68:19.
  32. Underhill J, Donaldson P, Bray G, et al. Susceptibility to primary biliary cirrhosis is associated with the HLA-DR8-DQB1*0402 haplotype. Hepatology 1992; 16:1404.
  33. Gregory WL, Mehal W, Dunn AN, et al. Primary biliary cirrhosis: contribution of HLA class II allele DR8. Q J Med 1993; 86:393.
  34. Qin B, Wang J, Chen J, et al. Association of human leukocyte antigen class II with susceptibility to primary biliary cirrhosis: a systematic review and meta-analysis. PLoS One 2013; 8:e79580.
  35. Mella JG, Roschmann E, Maier KP, Volk BA. Association of primary biliary cirrhosis with the allele HLA-DPB1*0301 in a German population. Hepatology 1995; 21:398.
  36. Underhill JA, Donaldson PT, Doherty DG, et al. HLA DPB polymorphism in primary sclerosing cholangitis and primary biliary cirrhosis. Hepatology 1995; 21:959.
  37. Miller KB, Sepersky RA, Brown KM, et al. Genetic abnormalities of immunoregulation in primary biliary cirrhosis. Am J Med 1983; 75:75.
  38. Juran BD, Atkinson EJ, Schlicht EM, et al. Primary biliary cirrhosis is associated with a genetic variant in the 3' flanking region of the CTLA4 gene. Gastroenterology 2008; 135:1200.
  39. Pulickal AS, Hambleton S, Callaghan MJ, et al. Biliary cirrhosis in a child with inherited interleukin-12 deficiency. J Trop Pediatr 2008; 54:269.
  40. Hirschfield GM, Liu X, Han Y, et al. Variants at IRF5-TNPO3, 17q12-21 and MMEL1 are associated with primary biliary cirrhosis. Nat Genet 2010; 42:655.
  41. Liu X, Invernizzi P, Lu Y, et al. Genome-wide meta-analyses identify three loci associated with primary biliary cirrhosis. Nat Genet 2010; 42:658.
  42. Bogdanos DP, Smyk DS, Rigopoulou EI, et al. Twin studies in autoimmune disease: genetics, gender and environment. J Autoimmun 2012; 38:J156.
  43. Selmi C, Mayo MJ, Bach N, et al. Primary biliary cirrhosis in monozygotic and dizygotic twins: genetics, epigenetics, and environment. Gastroenterology 2004; 127:485.
  44. Kaplan MM, Bianchi DW. Primary biliary cirrhosis: for want of an X chromosome? Lancet 2004; 363:505.
  45. Invernizzi P, Miozzo M, Battezzati PM, et al. Frequency of monosomy X in women with primary biliary cirrhosis. Lancet 2004; 363:533.
  46. Marzorati S, Lleo A, Carbone M, et al. The epigenetics of PBC: The link between genetic susceptibility and environment. Clin Res Hepatol Gastroenterol 2016; 40:650.
  47. Kikuchi K, Lian ZX, Yang GX, et al. Bacterial CpG induces hyper-IgM production in CD27(+) memory B cells in primary biliary cirrhosis. Gastroenterology 2005; 128:304.
  48. Zhang J, Zhang W, Leung PS, et al. Ongoing activation of autoantigen-specific B cells in primary biliary cirrhosis. Hepatology 2014; 60:1708.
  49. Selmi C, Ross SR, Ansari AA, et al. Lack of immunological or molecular evidence for a role of mouse mammary tumor retrovirus in primary biliary cirrhosis. Gastroenterology 2004; 127:493.
  50. Mason AL, Farr GH, Xu L, et al. Pilot studies of single and combination antiretroviral therapy in patients with primary biliary cirrhosis. Am J Gastroenterol 2004; 99:2348.
  51. Harada K, Tsuneyama K, Sudo Y, et al. Molecular identification of bacterial 16S ribosomal RNA gene in liver tissue of primary biliary cirrhosis: is Propionibacterium acnes involved in granuloma formation? Hepatology 2001; 33:530.
  52. Abdulkarim AS, Petrovic LM, Kim WR, et al. Primary biliary cirrhosis: an infectious disease caused by Chlamydia pneumoniae? J Hepatol 2004; 40:380.
  53. Leung PS, Park KO, Gershwin ME. Is there a relation between chlamydia infection and primary biliary cirrhosis? (abstract). Hepatology 2002; 36:388A.
  54. Maddala YK, Jorgensen RA, Angulo P, Lindor KD. Open-label pilot study of tetracycline in the treatment of primary biliary cirrhosis. Am J Gastroenterol 2004; 99:566.
  55. Ohno N, Ota Y, Hatakeyama S, et al. A patient with E. coli-induced pyelonephritis and sepsis who transiently exhibited symptoms associated with primary biliary cirrhosis. Intern Med 2003; 42:1144.
  56. Bogdanos DP, Baum H, Grasso A, et al. Microbial mimics are major targets of crossreactivity with human pyruvate dehydrogenase in primary biliary cirrhosis. J Hepatol 2004; 40:31.
  57. Shimoda S, Nakamura M, Shigematsu H, et al. Mimicry peptides of human PDC-E2 163-176 peptide, the immunodominant T-cell epitope of primary biliary cirrhosis. Hepatology 2000; 31:1212.
  58. O'Donohue J, Workman MR, Rolando N, et al. Urinary tract infections in primary biliary cirrhosis and other chronic liver diseases. Eur J Clin Microbiol Infect Dis 1997; 16:743.
  59. Butler P, Hamilton-Miller J, Baum H, Burroughs AK. Detection of M2 antibodies in patients with recurrent urinary tract infection using an ELISA and purified PBC specific antigens. Evidence for a molecular mimicry mechanism in the pathogenesis of primary biliary cirrhosis? Biochem Mol Biol Int 1995; 35:473.
  60. Yang Y, Choi J, Chen Y, et al. E. coli and the etiology of human PBC: Antimitochondrial antibodies and spreading determinants. Hepatology 2022; 75:266.
  61. Selmi C, Balkwill DL, Invernizzi P, et al. Patients with primary biliary cirrhosis react against a ubiquitous xenobiotic-metabolizing bacterium. Hepatology 2003; 38:1250.
  62. Olafsson S, Gudjonsson H, Selmi C, et al. Antimitochondrial antibodies and reactivity to N. aromaticivorans proteins in Icelandic patients with primary biliary cirrhosis and their relatives. Am J Gastroenterol 2004; 99:2143.
  63. Kaplan MM. Novosphingobium aromaticivorans: a potential initiator of primary biliary cirrhosis. Am J Gastroenterol 2004; 99:2147.
  64. Bogdanos D, Pusl T, Rust C, et al. Primary biliary cirrhosis following Lactobacillus vaccination for recurrent vaginitis. J Hepatol 2008; 49:466.
  65. Long SA, Quan C, Van de Water J, et al. Immunoreactivity of organic mimeotopes of the E2 component of pyruvate dehydrogenase: connecting xenobiotics with primary biliary cirrhosis. J Immunol 2001; 167:2956.
  66. Bruggraber SF, Leung PS, Amano K, et al. Autoreactivity to lipoate and a conjugated form of lipoate in primary biliary cirrhosis. Gastroenterology 2003; 125:1705.
  67. Leung PS, Quan C, Park O, et al. Immunization with a xenobiotic 6-bromohexanoate bovine serum albumin conjugate induces antimitochondrial antibodies. J Immunol 2003; 170:5326.
  68. Leung PS, Park O, Tsuneyama K, et al. Induction of primary biliary cirrhosis in guinea pigs following chemical xenobiotic immunization. J Immunol 2007; 179:2651.
  69. Wakabayashi K, Lian ZX, Leung PS, et al. 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.
  70. Coppel RL, McNeilage LJ, Surh CD, et al. Primary structure of the human M2 mitochondrial autoantigen of primary biliary cirrhosis: dihydrolipoamide acetyltransferase. Proc Natl Acad Sci U S A 1988; 85:7317.
  71. Van de Water J, Cooper A, Surh CD, et al. Detection of autoantibodies to recombinant mitochondrial proteins in patients with primary biliary cirrhosis. N Engl J Med 1989; 320:1377.
  72. Van de Water J, Fregeau D, Davis P, et al. Autoantibodies of primary biliary cirrhosis recognize dihydrolipoamide acetyltransferase and inhibit enzyme function. J Immunol 1988; 141:2321.
  73. Nishio A, Coppel R, Ishibashi H, Gershwin ME. The pyruvate dehydrogenase complex as a target autoantigen in primary biliary cirrhosis. Baillieres Best Pract Res Clin Gastroenterol 2000; 14:535.
  74. Shigematsu H, Shimoda S, Nakamura M, et al. Fine specificity of T cells reactive to human PDC-E2 163-176 peptide, the immunodominant autoantigen in primary biliary cirrhosis: implications for molecular mimicry and cross-recognition among mitochondrial autoantigens. Hepatology 2000; 32:901.
  75. Davis PA, Leung P, Manns M, et al. M4 and M9 antibodies in the overlap syndrome of primary biliary cirrhosis and chronic active hepatitis: epitopes or epiphenomena? Hepatology 1992; 16:1128.
  76. Palmer JM, Yeaman SJ, Bassendine MF, James OF. M4 and M9 autoantigens in primary biliary cirrhosis--a negative study. J Hepatol 1993; 18:251.
  77. Miyakawa H, Tanaka A, Kikuchi K, et al. Detection of antimitochondrial autoantibodies in immunofluorescent AMA-negative patients with primary biliary cirrhosis using recombinant autoantigens. Hepatology 2001; 34:243.
  78. Kaplan MM. Primary biliary cirrhosis. N Engl J Med 1996; 335:1570.
  79. Van Norstrand MD, Malinchoc M, Lindor KD, et al. Quantitative measurement of autoantibodies to recombinant mitochondrial antigens in patients with primary biliary cirrhosis: relationship of levels of autoantibodies to disease progression. Hepatology 1997; 25:6.
  80. Invernizzi P, Crosignani A, Battezzati PM, et al. Comparison of the clinical features and clinical course of antimitochondrial antibody-positive and -negative primary biliary cirrhosis. Hepatology 1997; 25:1090.
  81. Krams SM, Surh CD, Coppel RL, et al. Immunization of experimental animals with dihydrolipoamide acetyltransferase, as a purified recombinant polypeptide, generates mitochondrial antibodies but not primary biliary cirrhosis. Hepatology 1989; 9:411.
  82. Kim WR, Poterucha JJ, Jorgensen RA, et al. Does antimitochondrial antibody status affect response to treatment in patients with primary biliary cirrhosis? Outcomes of ursodeoxycholic acid therapy and liver transplantation. Hepatology 1997; 26:22.
  83. Colucci G, Schaffner F, Paronetto F. In situ characterization of the cell-surface antigens of the mononuclear cell infiltrate and bile duct epithelium in primary biliary cirrhosis. Clin Immunol Immunopathol 1986; 41:35.
  84. Shimoda S, Van de Water J, Ansari A, et al. Identification and precursor frequency analysis of a common T cell epitope motif in mitochondrial autoantigens in primary biliary cirrhosis. J Clin Invest 1998; 102:1831.
  85. Migliaccio C, Van de Water J, Ansari AA, et al. Heterogeneous response of antimitochondrial autoantibodies and bile duct apical staining monoclonal antibodies to pyruvate dehydrogenase complex E2: the molecule versus the mimic. Hepatology 2001; 33:792.
  86. Kita H, Matsumura S, He XS, et al. Quantitative and functional analysis of PDC-E2-specific autoreactive cytotoxic T lymphocytes in primary biliary cirrhosis. J Clin Invest 2002; 109:1231.
  87. Kita H, Naidenko OV, Kronenberg M, et al. Quantitation and phenotypic analysis of natural killer T cells in primary biliary cirrhosis using a human CD1d tetramer. Gastroenterology 2002; 123:1031.
  88. Van de Water J, Gershwin ME, Leung P, et al. The autoepitope of the 74-kD mitochondrial autoantigen of primary biliary cirrhosis corresponds to the functional site of dihydrolipoamide acetyltransferase. J Exp Med 1988; 167:1791.
  89. Fregeau DR, Prindiville T, Coppel RL, et al. Inhibition of alpha-ketoglutarate dehydrogenase activity by a distinct population of autoantibodies recognizing dihydrolipoamide succinyltransferase in primary biliary cirrhosis. Hepatology 1990; 11:975.
  90. Van de Water J, Ansari A, Prindiville T, et al. Heterogeneity of autoreactive T cell clones specific for the E2 component of the pyruvate dehydrogenase complex in primary biliary cirrhosis. J Exp Med 1995; 181:723.
  91. Shimoda S, Nakamura M, Ishibashi H, et al. HLA DRB4 0101-restricted immunodominant T cell autoepitope of pyruvate dehydrogenase complex in primary biliary cirrhosis: evidence of molecular mimicry in human autoimmune diseases. J Exp Med 1995; 181:1835.
  92. Kita H, Lian ZX, Van de Water J, et al. Identification of HLA-A2-restricted CD8(+) cytotoxic T cell responses in primary biliary cirrhosis: T cell activation is augmented by immune complexes cross-presented by dendritic cells. J Exp Med 2002; 195:113.
  93. Kita H, Matsumura S, He XS, et al. Analysis of TCR antagonism and molecular mimicry of an HLA-A0201-restricted CTL epitope in primary biliary cirrhosis. Hepatology 2002; 36:918.
  94. Malmborg AC, Shultz DB, Luton F, et al. Penetration and co-localization in MDCK cell mitochondria of IgA derived from patients with primary biliary cirrhosis. J Autoimmun 1998; 11:573.
  95. Portmann B, Popper H, Neuberger J, Williams R. Sequential and diagnostic features in primary biliary cirrhosis based on serial histologic study in 209 patients. Gastroenterology 1985; 88:1777.
  96. Kneppelhout JC, Mulder CJ, van Berge Henegouwen GP, et al. Ursodeoxycholic acid treatment in primary biliary cirrhosis with the emphasis on late stage disease. Neth J Med 1992; 41:11.
  97. Heuman DM, Pandak WM, Hylemon PB, Vlahcevic ZR. Conjugates of ursodeoxycholate protect against cytotoxicity of more hydrophobic bile salts: in vitro studies in rat hepatocytes and human erythrocytes. Hepatology 1991; 14:920.
  98. Schlaeger R, Haux P, Kattermann R. Studies on the mechanism of the increase in serum alkaline phosphatase activity in cholestasis: significance of the hepatic bile acid concentration for the leakage of alkaline phosphatase from rat liver. Enzyme 1982; 28:3.
  99. Montano-Loza AJ, Hansen BE, Corpechot C, et al. Factors Associated With Recurrence of Primary Biliary Cholangitis After Liver Transplantation and Effects on Graft and Patient Survival. Gastroenterology 2019; 156:96.
  100. Odin JA, Huebert RC, Casciola-Rosen L, et al. Bcl-2-dependent oxidation of pyruvate dehydrogenase-E2, a primary biliary cirrhosis autoantigen, during apoptosis. J Clin Invest 2001; 108:223.
  101. Lleo A, Bowlus CL, Yang GX, et al. Biliary apotopes and anti-mitochondrial antibodies activate innate immune responses in primary biliary cirrhosis. Hepatology 2010; 52:987.
  102. Lleo A, Selmi C, Invernizzi P, et al. Apotopes and the biliary specificity of primary biliary cirrhosis. Hepatology 2009; 49:871.
  103. Van de Water J, Ansari AA, Surh CD, et al. Evidence for the targeting by 2-oxo-dehydrogenase enzymes in the T cell response of primary biliary cirrhosis. J Immunol 1991; 146:89.
  104. Jones DE, Palmer JM, James OF, et al. T-cell responses to the components of pyruvate dehydrogenase complex in primary biliary cirrhosis. Hepatology 1995; 21:995.
  105. Van de Water J, Turchany J, Leung PS, et al. Molecular mimicry in primary biliary cirrhosis. Evidence for biliary epithelial expression of a molecule cross-reactive with pyruvate dehydrogenase complex-E2. J Clin Invest 1993; 91:2653.
  106. Tsuneyama K, Van de Water J, Leung PS, et al. Abnormal expression of the E2 component of the pyruvate dehydrogenase complex on the luminal surface of biliary epithelium occurs before major histocompatibility complex class II and BB1/B7 expression. Hepatology 1995; 21:1031.
  107. Sasaki M, Sato Y, Nakanuma Y. An impaired biliary bicarbonate umbrella may be involved in dysregulated autophagy in primary biliary cholangitis. Lab Invest 2018; 98:745.
  108. Nelson JL, Furst DE, Maloney S, et al. Microchimerism and HLA-compatible relationships of pregnancy in scleroderma. Lancet 1998; 351:559.
  109. Corpechot C, Barbu V, Chazouillères O, Poupon R. Fetal microchimerism in primary biliary cirrhosis. J Hepatol 2000; 33:696.
  110. Fanning PA, Jonsson JR, Clouston AD, et al. Detection of male DNA in the liver of female patients with primary biliary cirrhosis. J Hepatol 2000; 33:690.
  111. Rubbia-Brandt L, Philippeaux MM, Chavez S, et al. FISH for Y chromosome in women with primary biliary cirrhosis: lack of evidence for leukocyte microchimerism. Hepatology 1999; 30:821.
  112. Tanaka A, Lindor K, Gish R, et al. Fetal microchimerism alone does not contribute to the induction of primary biliary cirrhosis. Hepatology 1999; 30:833.
  113. Jones DE. Fetal microchimerism: an aetiological factor in primary biliary cirrhosis? J Hepatol 2000; 33:834.
Topic 3616 Version 27.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟