metricas
covid
Buscar en
Annals of Hepatology
Toda la web
Inicio Annals of Hepatology Smoking as a risk factor for autoimmune liver disease: what we can learn from pr...
Journal Information
Vol. 11. Issue 1.
Pages 7-14 (January - February 2012)
Share
Share
Download PDF
More article options
Visits
2498
Vol. 11. Issue 1.
Pages 7-14 (January - February 2012)
Open Access
Smoking as a risk factor for autoimmune liver disease: what we can learn from primary biliary cirrhosis
Visits
2498
Daniel S. Smyk*, Eirini I. Rigopoulou**, Luigi Muratori***, Andrew K. Burroughs****, Dimitrios P. Bogdanos
,
Corresponding author
dimitrios.bogdanos@kcl.ac

Correspondence and reprint request:
* Institute of Liver Studies, King’s College London School of Medicine at King’s College Hospital, Denmark Hill Campus, London SE5 9RS, UK
** Department of Medicine, University of Thessaly Medical School, Thessaly, Mezourlo, Larissa 41110, Greece
*** Department of Clinical Medicine, Alma Mater Studiorum-University of Bologna, Policlinico Sant'Orsola-Malpighi, Padiglione 11, Bologna, Italy
**** The Sheila Sherlock Liver Centre, and University Department of Surgery, Royal Free Hospital, London NW3 2QG, UK
This item has received

Under a Creative Commons license
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Tables (1)
Table 1.. Evidence in support and against the role of cigarette smoke in the pathogenesis of primary biliary cirrhosis (PBC).
Abstract

Primary biliary cirrhosis (PBC) is a cholestatic liver disease characterised by the immune-mediated destruction of biliary epithelial cells in small intrahepatic bile ducts. The disease is characterised by circulating anti-mitochondrial antibodies (AMA) as well as disease specific anti-nuclear antibodies (ANA), cholestatic liver biochemistry, and characteristic histology. The disease primarily affects middle-aged females, and its incidence is apparently increasing worldwide. Epidemiological studies have indicated several risk factors for the development of PBC, with family history of PBC, recurrent urinary tract infection, and smoking being the most widely cited. Smoking has been implicated as a risk factor in several autoimmune diseases, including the liver, by complex mechanisms involving the endocrine and immunological systems to name a few. Studies of smoking in liver disease have also shown that smoking may progress the disease towards fibrosis and subsequent cirrhosis. This review will examine the literature surrounding smoking as a risk factor for PBC, as well as a potential factor in the progression of fibrosis in PBC patients.

Key words:
Autoimmunity
Autoimmune disease
Cigarette
Liver
Prevention
Smoking
Full Text
Introduction

Primary biliary cirrhosis (PBC) is characterised by the autoimmune destruction of small intrahepatic bile ducts, with fibrosis progressing to cirrhosis and eventual liver failure.1-6 Patient presentation varies, with some patients being asymptomatic with liver function tests indicating cholestasis.2,7-10 Symptomatic patients typically present with pruritus, fatigue and arthralgias,2,10,11 with more severe symptoms being related to portal hypertension, including ascites, jaundice, or variceal bleeding.2,10,11 The diagnosis of PBC is based on biochemical markers of choles-tasis, histological features of PBC, and the presence of disease specific anti-mitochondrial antibodies (AMA) and/or disease-specific anti-nuclear antibodies (ANA).1-3,7-10,12-31 AMA is pathognomonic for PBC with the presence of the disease is questioned in their absence' and it is commonly the case that AMA is predictive of future disease development in asymptomatic individuals.1-3,7-10,12-22,26,28,32 These autoantibodies also show a higher prevalence among family members of patients with PBC, and AMA-positive individuals are at risk of developing PBC.33 AMA specific to PBC are directed against components of the 2-oxo-acid dehydrogenase complexes (previously known as M2 antigens), primarily the E2 subunit of the pyruvate dehydrogenase complex, or PDC-E2.2,8,9,13,25,34-38 Medical treatment of PBC includes urseodeoxycholic acid, which frequently leads to a good biochemical response and decreased indices of cholestasis in those diagnosed in the early stages of the disease, or liver transplantation in more severe cases.1-3,5,10,39-46

The aetiology of PBC is not fully understood, but it appears that a variety of factors including genetics, immunological factors, as well as exposure to xenobiotic and infectious agents may account for the development of the disease.17,47-61 Genetic and genome wide association studies (GWAS) have demonstrated associations with several HLA and non-HLA regions.54 Associated regions include HLA-DRB1, DQA1, DQB1, and DQA2, and non-HLA regions such as IRF5, SPIB, the IKZF3-ORMDL3 of chromosome 17q12-21, IL12A and IL12RB.54,62 IRF5-TNPO3, 17q12-21 and MMEL1 have been found to be associated with primary biliary cirrhosis in other studies.50 The most recent GWAS on PBC included 1,840 PBC patients and 5,163 controls from the UK, identified twelve new susceptibility loci including new candidate genes STAT4, DENND1B, CD80, IL7R, CXCR5, TNFRSF1A, CLEC16A and NFKB1.58

Infectious agents have been implicated, largely through mechanisms such as molecular mimicry and cross reactivity.4,7,9,12,36,63-79 Other compounds including chemicals have also been implicated in the development of PBC, including contamination of air, water, and soil.80,81 Several studies have consistently indicated that several risk factors for PBC development, including recurrent urinary tract infection and smoking.82-87 Smoking has been implicated in several autoimmune diseases, and its effects may be multifactorial, involving tissue damage and apoptosis, inflammation, and anti-oestrogenic effects.88-101 This review will examine the role of smoking as a risk factor for the development of PBC, as well as the implication of smoking in regards to liver fibrosis (Table 1).

Table 1..

Evidence in support and against the role of cigarette smoke in the pathogenesis of primary biliary cirrhosis (PBC).

Evidence in support of smoking in the pathogenesis of PBC  Evidence against smoking in the pathogenesis of PBC 
Associations found in all large epidemiological studies  Lack of apparent mechanism for the induction of biliary pathology 
Advanced fibrosis associated with a significant smoking history  Variability between epidemiological reports 
Increased risk of advanced fibrosis with increased number of pack years  Liver enzyme levels did not differ between smokers and non-smokers 
Significant hepatic inflammatory activity seen in smokers vs. non-smokers  Interlobular bile duct paucity more severe in non-smokers 
Increased IFN-γ and IL-10 in smokers, reflecting an increased Th1 response  No difference in immunoglobulin or autoantibody levels between smokers and non-smokers 
Chemicals in cigarette smoke also implicated in PBC clusters   
Epidemiological Studies

Over the past decade, epidemiological studies on PBC have identified multiple risk factors for PBC development, ranging from familial and past surgical history, to nail polish use and oestrogen deficiency.48,82,84-86 Several risk factors, however, havebeen indicated in virtually all major epidemiologitcal studies, namely urinary tract infection, family history and smoking.82,84-86 Howel, et al. conducted a population-based case-control study of PBC in the North East of England, using postal questionnaires covering medical history and lifestyle sent to 100 PBC patients and 223 controls.102 An unexpected association was found with smoking, where 76% of PBC and 57% of controls had ever smoked.102 Parikh-Patel, et al.85 conducted a study on 199 PBC patients, 171 of their siblings, and 141 of their friends as controls. They reported that 66.2% of PBC patients had a history of smoking, compared to 62.2% of their siblings and 49.9% of their friends.85 Prince, et al. also found an association between PBC and smoking in their study, comprised of two PBC groups: one from an epidemiological study consisting of 318 cases, and the other from a PBC support group consisting of 2,258 cases.86 The odds ratio (OR) for those who had ever smoked was 1.63 in the epidemiological group, and 1.57 in the support group.86 Multivariate analysis demonstrated an adjusted OR of 1.6 compared to the epidemiological cases, and 1.5 compared to the support group cases.86

One of the largest, most comprehensive epidemiological studies on PBC was conducted by Gershwin, et al.,84 involving 1,032 PBC patients from 23 different tertiary care facilities, and 1,041 controls. Controls were selected from random digit dialling, and both controls and PBC patients were administered a telephone questionnaire covering lifestyle and medical history.84 Past smoking was found to be significantly associated with the development of PBC, with 60% of patients and 54% of controls having smoked more than 100 cigarettes in their lifetime.84 Strangely, this association was related only to past smoking, as only 16% of PBC patients but 32% of controls currently smoking at the time the study was conducted.84 This may be due to the cessation of smoking by PBC patients at the time of diagnosis, as Parikh-Patel, et al. found that 21.4% of patients stopped smoking at the time they were diagnosed with PBC.85 Former smokers comprised 46.5% of the PBC group, compared to 36.6% of siblings and 34.8% of friends.85 A higher percentage of individuals who had never smoked was found in controls groups, with 51.1% of friends and 37.8% of siblings having never smoked, compared to 33.8% of PBC patients.85

The above studies have indicated an association with past smoking and PBC. An epidemiological analysis conducted by Corpechot, et al.82 went further and examined the role of active and passive smoking. That study involved 222 PBC patients and 509 controls, all of whom were administered a questionnaire which included details on exposure to cigarette smoke.82 A history of cigarette smoke was defined as having consumed more than 100 cigarettes in a lifetime, and active smoking was defined as daily smoking for the past 6 months.82 Active and/or passive smoking was found to be associated with PBC, with 45% of patients, and 19% of controls reporting either an active or passive smoking history.82 Active smoking was reported in 20% of PBC patients and 13% of controls.82 Persistent exposure to cigarette smoke was reported in 39% of PBC cases and 16% of controls.82 Finally, a meta-analysis of five existing studies demonstrated that smoking, family history of PBC, and a personal history of urinary tract infection were strongly associated with PBC.87 Those authors note that all studies included in the analysis were based on a Caucasian population, and that studies are needed in different populations to determine if there are varied risk factors depending on ethnic grouping.87 As well, the cumulative effect of a number of potential risk factors is not clear, and has not been addressed in any of the above studies. For example, it is unknown as to how many patients with a history of urinary tract infection or family history of PBC, also had a significant smoking history. An analysis of these relationships may shed some light on the additive effects of a variety of triggers.

Smoking and Fibrosis In PBC

Epidemiological studies indicate that smoking, or at least a past history of smoking, is associated with PBC. Emerging evidence also appears to indicate that exposure to cigarette smoke may also have an impact on the progression of the disease towards fibrosis.83,90,93,95,99,101,103-110 Previous studies in hepatitis C and alcoholic liver disease have indicated that smoking may impact the severity of fibrosis.99,101,106,108,111 A study by Zein, et al.109 attempted to determine the relationship between smoking and the severity of liver fibrosis at presentation in patients with PBC. Over a seven year period, 97 patients were retrospectively identified, and the cumulative number of cigarette packs smoked per year (pack-years) was calculated for each patient.109 Advanced fibrosis (stage 3 or 4) was found to be associated with increased lifetime tobacco consumption, being greater than or equal to 10 pack years.109 The association remained significant after adjusting for age, gender and alcohol intake, and cross-validation of 172 PBC patients confirmed these findings.109

A recent study has also demonstrated an association between smoking and advanced fibrosis, as well as biochemical and immunological changes.83 A total of 223 PBC patients were interviewed regarding their smoking habits using a questionnaire, and histological data from presentation was available from 164 patients.83 A significant smoking history was reported by 26% of patients (current or past history of smoking five or more cigarettes per day), 11% were active smokers, and 41% were non-smokers who had been exposed to cigarette smoke consistently, either at home or at work.83 Advanced fibrosis (stage 3 or 4) was found in 37% of current smokers and 33% of past smokers, compared to only 16% of those who had never smoked.83 In fact, each pack year of increase in smoking intensity was associated with a 5% increase in the likelihood of advanced fibrosis.83 Florid destructive cholangitis was found to be more prevalent in past smokers (63%) followed by current smokers (61%) and non-smokers (57%).83 Significant inflammatory activity was seen in 33% of smokers vs. 32% of non-smokers, and strangely, interlobular bile duct paucity was less prevalent in current and past smokers (31 and 30% respectively) vs. non-smokers (54%).83 Liver enzyme activities (ALP, GGT, AST, ALT) and concentrations of serum total bilirubin and albumin did not differ significantly between smoking and non-smoking groups.83 No differences were observed in serum immunoglobulin levels, or in autoantibody titres.83 Although further studies are needed to determine the mechanism underlying smoking as a factor for progressive fibrosis, it is recommended that patients with liver disease be encouraged to stop smoking.83,103,112

The Immunomodulatory Properties of Smoking: A Pathway to Autoimmunity?

The effects of smoking on the immune system are diverse, and the roles those changes play in autoimmunity have not been well defined.113 Alterations in cytokine production, such as IL-13, and the subsequent balance of T lymphocyte subtypes have been implicated in epidemiological studies.84-86 Gershwin, et al. note the possible effects of tobacco smoke on the Th1 cytokine response, which has found some support in regards to COPD, another autoimmune disease.84,98,100 Whetzel, et al.100 measured Th1 and Th2 immune responses via IFN-γ an IL-10 in 20 healthy non-smokers and 19 smokers aged 19-41 years. Smokers had increased IFN-γ and IL-10, even after 24 h of smoking abstinence.100 IFN-γ levels were higher in female smokers, but no age or sex difference was noted with IL-10.100 That study and others have demonstrated a predominance of the Th1 cell line in COPD,100,114-117 and that the predominance of these cells may occur due to cigarette smoking. This is of interest in PBC, given that Th1 cells are the more prominent T-cell type of lymphoid infiltrates seen in PBC.118

Chemical compounds found in cigarette smoke have also been implicated in clusters of PBC. Ala, et al.119 report a cluster of PBC patients near a super-fund toxic waste site contaminated by volatile aromatic hydrocarbons. As there was no apparent contamination of groundwater, it was suggested that air contamination by chlorinated hydrocarbons, such as benzene, may be a potential factor in the development of PBC.119 It is also possible that benzene-containing cigarette smoke may also be associated with PBC.82,84,85,119 Immunological changes in response to exposure to chemical constituents of cigarette smoke, warrant further investigation on the experimental level.

Conclusion

The deleterious effects of smoking are now well known, and appear to be involved in the pathogenesis and progression of many diseases, including autoimmune diseases. The mechanisms involved are complex, and involve alterations in immunological and endocrine function, as well as the production of free radicals and oxidative stress. Epidemiological studies have strongly implicated smoking as a risk factor for PBC, and recent studies also indicate that smoking may also increase the risk of fibrosis in PBC patients. Although further studies are needed to confirm and clarify those studies, it is clear that smoking should be strongly discouraged among patients with PBC and other chronic liver diseases. Although it is not possible to 'un-do' the damage that smoking may have caused in patients at presentation, it may not be too late to educate patients on the possible effects of smoking on their cohabitating relatives. A family history of PBC has been shown to increase the risk of PBC, and therefore, action must be taken to reduce the risk of relatives developing PBC, such as by screening, but also in regards to harm reduction, such as reduction of smoking in the household. Although this may not prevent another diagnosis of PBC, it may have implications as to the severity of the disease at presentation, and hence patient outcome.

Abbreviations

  • AMA: Anti-mitochondrial antibodies.

  • ANA: anti-nuclear antibodies.

  • PBC: primary biliary cirrhosis.

  • PDC: pyruvate dehydrogenase complex.

References
[1.]
Hohenester S., Oude-Elferink R.P., Beuers U..
Primary biliary cirrhosis.
Semin Immunopathol, 31 (2009), pp. 283-307
[2.]
Kaplan M.M., Gershwin M.E..
Primary biliary cirrhosis.
N Engl J Med, 353 (2005), pp. 1261-1273
[3.]
Lindor K.D., Gershwin M.E., Poupon R., Kaplan M., Bergasa N.V., Heathcote E.J..
Primary biliary cirrhosis.
Hepatology, 50 (2009), pp. 291-308
[4.]
Agmon-Levin N., Katz B.S., Shoenfeld Y..
Infection and primary biliary cirrhosis.
Isr Med Assoc J, 11 (2009), pp. 112-115
[5.]
Poupon R..
Primary biliary cirrhosis: a 2010 update.
J Hepatol, 52 (2010), pp. 745-758
[6.]
Poupon R., Chazouilleres O., Poupon R.E..
Chronic cholestatic diseases.
J Hepatol, 32 (2000), pp. 129-140
[7.]
Bogdanos D.P., Baum H., Vergani D..
Antimitochondrial and other autoantibodies.
Clin Liver Dis, 7 (2003), pp. 759-777
[8.]
Bogdanos D.P., Invernizzi P., Mackay I.R., Vergani D..
Autoimmune liver serology: current diagnostic and clinical challenges.
World J Gastroenterol, 14 (2008), pp. 3374-3387
[9.]
Bogdanos D.P., Komorowski L..
Disease-specific autoantibodies in primary biliary cirrhosis.
Clin Chim Acta, 412 (2011), pp. 502-512
[10.]
Neuberger J..
Primary biliary cirrhosis.
[11.]
Kumagi T., Heathcote E.J..
Primary biliary cirrhosis.
Orphanet J Rare Dis, 3 (2008), pp. 1
[12.]
Bogdanos D.P., Liaskos C., Pares A., Norman G., Rigopoulou E.I., Caballeria L., et al.
Anti-gp210 antibody mirrors disease severity in primary biliary cirrhosis.
Hepatology, 45 (2007), pp. 1583-1584
[13.]
Bogdanos D.P., Liaskos C., Rigopoulou E.I., Dalekos G.N..
Anti-mitochondrial antibodies in patients with systemic lupus erythematosus: revealing the unforeseen.
Clin Chim Acta, 373 (2006), pp. 183-184
[14.]
Bogdanos D.P., Pares A., Rodes J., Vergani D..
Primary biliary cirrhosis specific antinuclear antibodies in patients from Spain.
Am J Gastroenterol, 99 (2004), pp. 763-764
[15.]
Dahnrich C., Pares A., Caballeria L., Rosemann A., Schlumberger W., Probst C., et al.
New ELISA for detecting primary biliary cirrhosis-specific antimitochondrial antibodies.
Clin Chem, 55 (2009), pp. 978-985
[16.]
Gabeta S., Norman G.L., Liaskos C., Papamichalis P.A., Zografos T., Garagounis A., et al.
Diagnostic relevance and clinical significance of the new enhanced performance M2 (MIT3) ELISA for the detection of IgA and IgG antimitochondrial antibodies in primary biliary cirrhosis.
J Clin Immunol, 27 (2007), pp. 378-387
[17.]
Gershwin M.E., Mackay I.R..
Primary biliary cirrhosis: paradigm or paradox for autoimmunity.
Gastroenterology, 100 (1991), pp. 822-833
[18.]
Liu H., Norman G.L., Shums Z., Worman H.J., Krawitt E.L., Bizzaro N., et al.
PBC screen: an IgG/IgA dual isotype ELISA detecting multiple mitochondrial and nuclear autoantibodies specific for primary biliary cirrhosis.
J Autoimmun, 35 (2010), pp. 436-442
[19.]
Metcalf J.V., Mitchison H.C., Palmer J.M., Jones D.E., Bassendine M.F., James O.F..
Natural history of early primary biliary cirrhosis.
Lancet, 348 (1996), pp. 1399-1402
[20.]
Mitchison H.C., Bassendine M.F., Hendrick A., Bennett M.K., Bird G., Watson A.J., et al.
Positive antimitochondrial antibody but normal alkaline phosphatase: is this primary biliary cirrhosis?.
Hepatology, 6 (1986), pp. 1279-1284
[21.]
Rigopoulou E.I., Bogdanos D.P., Liaskos C., Koutsoumpas A., Baum H., Vergani D., et al.
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 (2007), pp. 118-121
[22.]
Rigopoulou E.I., Davies E.T., Pares A., Zachou K., Liaskos C., Bogdanos D.P., et al.
Prevalence and clinical significance of isotype specific antinuclear antibodies in primary biliary cirrhosis.
[23.]
Muratori L., Cassani F., Pappas G., Guidi M., Mele L., Lorenza V., et al.
The hepatitic/cholestatic “overlap” syndrome: an Italian experience.
Autoimmunity, 35 (2002), pp. 565-568
[24.]
Muratori P., Muratori L., Ferrari R., Cassani F., Bianchi G., Lenzi M., et al.
Characterization and clinical impact of antinuclear antibodies in primary biliary cirrhosis.
Am J Gastroenterol, 98 (2003), pp. 431-437
[25.]
Muratori P., Muratori L., Gershwin M.E., Czaja A.J., Pappas G., MacCariello S., et al.
‘True’ antimitochondrial antibody-negative primary biliary cirrhosis, low sensitivity of the routine assays, or both?.
Clin Exp Immunol, 135 (2004), pp. 154-158
[26.]
Granito A., Muratori P., Muratori L., Pappas G., Cassani F., Worthington J., et al.
Antibodies to SS-A/Ro-52kD and centromere in autoimmune liver disease: a clue to diagnosis and prognosis of primary biliary cirrhosis.
Aliment Pharmacol Ther, 26 (2007), pp. 831-838
[27.]
Muratori L., Granito A., Muratori P., Pappas G., Bianchi F.B..
Antimitochondrial antibodies and other antibodies in primary biliary cirrhosis: diagnostic and prognostic value.
Clin Liver Dis, 12 (2008), pp. 261-276
[28.]
Muratori P., Granito A., Pappas G., Muratori L., Lenzi M., Bianchi F.B..
Clinical and serological profile of primary biliary cirrhosis in young and elderly patients.
QJM, 101 (2008), pp. 505-506
[29.]
Muratori P., Granito A., Ferri S., Pappas G., Volta U., Menichella R., et al.
Multiple nuclear dots and rim-like/membranous IgG isotypes in primary biliary cirrhosis.
Autoimmunity, 42 (2009), pp. 224-227
[30.]
Muratori P., Granito A., Pappas G., Pendino G.M., Quarneti C., Cicola R., et al.
The serological profile of the autoimmune hepatitis/primary biliary cirrhosis overlap syndrome.
Am J Gastroenterol, 104 (2009), pp. 1420-1425
[31.]
Granito A., Yang W.H., Muratori L., Lim M.J., Nakajima A., Ferri S., et al.
PML nuclear body component Sp140 is a novel autoantigen in primary biliary cirrhosis.
Am J Gastroenterol, 105 (2010), pp. 125-131
[32.]
Vergani D., Bogdanos D.P..
Positive markers in AMA-negative PBC.
Am J Gastroenterol, 98 (2003), pp. 241-243
[33.]
Smyk D., Cholongitas E., Kriese S., Rigopoulou E.I., Bogdanos D.P..
Primary biliary cirrhosis: family stories.
Autoimmune Dis, 11 (2011), pp. 1-11
[34.]
Leung P.S., Coppel R.L., Ansari A., Munoz S., Gershwin M.E..
Antimitochondrial antibodies in primary biliary cirrhosis.
Semin Liver Dis, 17 (1997), pp. 61-69
[35.]
Rigopoulou E.I., Davies E.T., Bogdanos D.P., Liaskos C., Mytilinaiou M., Koukoulis G.K., et al.
Antimitochondrial antibodies of immunoglobulin G3 subclass are associated with a more severe disease course in primary biliary cirrhosis.
Liver Int, 27 (2007), pp. 1226-1231
[36.]
Butler P., Hamilton-Miller J., Baum H., Burroughs A.K..
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, 35 (1995), pp. 473-485
[37.]
Butler P., Valle F., Burroughs A.K..
Mitochondrial antigens and antibodies in primary biliary cirrhosis.
Postgrad Med J, 67 (1991), pp. 790-797
[38.]
Muratori L., Muratori P., Granito A., Ferrari R., Veronesi L., Lenzi M., et al.
The Western immunoblotting pattern of anti-mitochondrial antibodies is independent of the clinical expression of primary biliary cirrhosis.
Dig Liver Dis, 37 (2005), pp. 108-112
[39.]
Degott C., Zafrani E.S., Callard P., Balkau B., Poupon R.E., Poupon R..
Histopathological study of primary biliary cirrhosis and the effect of ursodeoxycholic acid treatment on histology progression.
Hepatology, 29 (1999), pp. 1007-1012
[40.]
Kaplan M.M., Poupon R..
Treatment with immunosuppressives in patients with primary biliary cirrhosis who fail to respond to ursodiol.
Hepatology, 50 (2009), pp. 652
[41.]
Poupon R..
Treatment of primary biliary cirrhosis with ursodeoxycholic Acid, budesonide and fibrates.
Dig Dis, 29 (2011), pp. 85-88
[42.]
Poupon R., Poupon R.E..
Treatment of primary biliary cirrhosis.
Baillieres Best Pract Res Clin Gastroenterol, 14 (2000), pp. 615-628
[43.]
Rabahi N., Chretien Y., Gaouar F., Wendum D., Serfaty L., Chazouilleres O., et al.
Triple therapy with ursodeoxycholic acid, budesonide and mycophenolate mofetil in patients with features of severe primary biliary cirrhosis not responding to ursodeoxycholic acid alone.
Gastroenterol Clin Biol, 34 (2010), pp. 283-287
[44.]
Burroughs A.K., Leandro G., Goulis J..
Ursodeooxycholic acid for primary biliary cirrhosis.
J Hepatol, 34 (2001), pp. 352-353
[45.]
Tsochatzis E.A., Gurusamy K.S., Gluud C., Burroughs A.K..
Ursodeoxycholic acid and primary biliary cirrhosis: EASL and AASLD guidelines.
J Hepatol, 51 (2009), pp. 1084-1085
[46.]
Manousou P., Arvaniti V., Tsochatzis E., Isgro G., Jones K., Shirling G., et al.
Primary biliary cirrhosis after liver transplantation: influence of immunosuppression and human leukocyte antigen locus disparity.
Liver Transpl, 16 (2010), pp. 64-73
[47.]
Mackay I.R., Whittingham S., Fida S., Myers M., Ikuno N., Gershwin M.E., et al.
The peculiar autoimmunity of primary biliary cirrhosis.
Immunol Rev, 174 (2000), pp. 226-237
[48.]
Gershwin M.E., Mackay I.R..
The causes of primary biliary cirrhosis: Convenient and inconvenient truths.
Hepatology, 47 (2008), pp. 737-745
[49.]
Hirschfield G.M., Heathcote E.J., Gershwin M.E..
Pathogenesis of cholestatic liver disease and therapeutic approaches.
Gastroenterology, 139 (2010), pp. 1481-1496
[50.]
Hirschfield G.M., Liu X., Han Y., Gorlov I.P., Lu Y., Xu C., et al.
Variants at IRF5-TNPO3, 17q12-21 and MMEL1 are associated with primary biliary cirrhosis.
Nat Genet, 42 (2010), pp. 655-657
[51.]
Invernizzi P..
Human leukocyte antigen in primary biliary cirrhosis: An old story now reviving.
Hepatology, 54 (2011), pp. 714-723
[52.]
Invernizzi P., Miozzo M., Battezzati P.M., Bianchi I., Grati F.R., Simoni G., et al.
Frequency of monosomy X in women with primary biliary cirrhosis.
[53.]
Invernizzi P., Selmi C., Poli F., Frison S., Floreani A., Alvaro D., et al.
Human leukocyte antigen polymorphisms in Italian primary biliary cirrhosis: a multicenter study of 664 patients and 1992 healthy controls.
Hepatology, 48 (2008), pp. 1906-1912
[54.]
Liu X., Invernizzi P., Lu Y., Kosoy R., Bianchi I., Podda M., et al.
Genome-wide meta-analyses identify three loci associated with primary biliary cirrhosis.
Nat Genet, 42 (2010), pp. 658-660
[55.]
Longhi M.S., Hussain M.J., Bogdanos D.P., Quaglia A., Mieli-Vergani G., Ma Y., et al.
Cytochrome P450IID6-specific CD8 T cell immune responses mirror disease activity in autoimmune hepatitis type 2.
Hepatology, 46 (2007), pp. 472-484
[56.]
Longhi M.S., Ma Y., Bogdanos D.P., Cheeseman P., Mieli-Vergani G., Vergani D..
Impairment of CD4(+)CD25(+) regulatory T-cells in autoimmune liver disease.
J Hepatol, 41 (2004), pp. 31-37
[57.]
Longhi M.S., Ma Y., Mitry R.R., Bogdanos D.P., Heneghan M., Cheeseman P., et al.
Effect of CD4+ CD25+ regulatory T-cells on CD8 T-cell function in patients with autoimmune hepatitis.
J Autoimmun, 25 (2005), pp. 63-71
[58.]
Mells G.F., Floyd J.A., Morley K.I., Cordell H.J., Franklin C.S., Shin S.Y., et al.
Genome-wide association study identifies 12 new susceptibility loci for primary biliary cirrhosis.
Nat Genet, 43 (2011), pp. 329-332
[59.]
Vergani D., Bogdanos D.P., Baum H..
Unusual suspects in primary biliary cirrhosis.
Hepatology, 39 (2004), pp. 38-41
[60.]
Selmi C., Invernizzi P., Keeffe E.B., Coppel R.L., Podda M., Rossaro L., et al.
Epidemiology and pathogenesis of primary biliary cirrhosis.
J Clin Gastroenterol, 38 (2004), pp. 264-271
[61.]
Watanabe N., Takashimizu S., Shiraishi K., Kagawa T., Nishizaki Y., Mine T., et al.
Primary biliary cirrhosis with multinucleated hepatocellular giant cells: implications for pathogenesis of primary biliary cirrhosis.
Eur J Gastroenterol Hepatol, 18 (2006), pp. 1023-1027
[62.]
Hirschfield G.M., Liu X., Xu C., Lu Y., Xie G., Gu X., et al.
Primary biliary cirrhosis associated with HLA, IL12A, and IL12RB2 variants.
N Engl J Med, 360 (2009), pp. 2544-2555
[63.]
Bogdanos D.P., Baum H., Butler P., Rigopoulou E.I., Davies E.T., Ma Y., et al.
Association between the primary biliary cirrhosis specific anti-sp100 antibodies and recurrent urinary tract infection.
Dig Liver Dis, 35 (2003), pp. 801-805
[64.]
Bogdanos D.P., Baum H., Grasso A., Okamoto M., Butler P., Ma Y., et al.
Microbial mimics are major targets of crossreactivity with human pyruvate dehydrogenase in primary biliary cirrhosis.
J Hepatol, 40 (2004), pp. 31-39
[65.]
Bogdanos D.P., Baum H., Gunsar F., Arioli D., Polymeros D., Ma Y., et al.
Extensive homology between the major immunodominant mitochondrial antigen in primary biliary cirrhosis and Helicobacter pylori does not lead to immunological cross-reactivity.
Scand J Gastroenterol, 39 (2004), pp. 981-987
[66.]
Bogdanos D.P., Baum H., Sharma U.C., Grasso A., Ma Y., Burroughs A.K., et al.
Antibodies against homologous microbial caseinolytic proteases P characterise primary biliary cirrhosis.
J Hepatol, 36 (2002), pp. 14-21
[67.]
Bogdanos D.P., Baum H., Vergani D., Burroughs A.K..
The role of E. coli infection in the pathogenesis of primary biliary cirrhosis.
Dis Markers, 29 (2010), pp. 301-311
[68.]
Bogdanos D.P., Choudhuri K., Vergani D..
Molecular mimicry and autoimmune liver disease: virtuous intentions, malign consequences.
Liver, 21 (2001), pp. 225-232
[69.]
Bogdanos D.P., Pares A., Baum H., Caballeria L., Rigopoulou E.I., Ma Y., et al.
Disease-specific cross-reactivity between mimicking peptides of heat shock protein of Mycobacterium gordonae and dominant epitope of E2 subunit of pyruvate dehydrogenase is common in Spanish but not British patients with primary biliary cirrhosis.
J Autoimmun, 22 (2004), pp. 353-362
[70.]
Bogdanos D.P., Mieli-Vergani G., Vergani D..
Autoantibodies and their antigens in autoimmune hepatitis.
Semin Liver Dis, 29 (2009), pp. 241-253
[71.]
Bogdanos D.P., Gilbert D., Bianchi I., Leoni S., Mitry R.R., Ma Y., et al.
Antibodies to soluble liver antigen and alpha-enolase in patients with autoimmune hepatitis.
J Autoimmune Dis, 1 (2004), pp. 4
[72.]
Baum H..
Mitochondrial antigens, molecular mimicry and autoimmune disease.
Biochim Biophys Acta, 1271 (1995), pp. 111-121
[73.]
Burroughs A.K., Butler P., Sternberg M.J., Baum H..
Molecular mimicry in liver disease.
Nature, 358 (1992), pp. 377-378
[74.]
Burroughs A.K., Rosenstein I.J., Epstein O., Hamilton-Miller J.M., Brumfitt W., Sherlock S..
Bacteriuria and primary biliary cirrhosis.
Gut, 25 (1984), pp. 133-137
[75.]
Butler P., Hamilton-Miller J.M., McIntyre N., Burroughs A.K..
Natural history of bacteriuria in women with primary biliary cirrhosis and the effect of antimicrobial therapy in symptomatic and asymptomatic groups.
Gut, 36 (1995), pp. 931-934
[76.]
Butler P., Valle F., Hamilton-Miller J.M., Brumfitt W., Baum H., Burroughs A.K..
M2 mitochondrial antibodies and urinary rough mutant bacteria in patients with primary biliary cirrhosis and in patients with recurrent bacteriuria.
J Hepatol, 17 (1993), pp. 408-414
[77.]
Burroughs A.K., Biagini M., McCormick P.A., Rolles K..
Liver transplantation and primary biliary cirrhosis.
Postgrad Med J, 65 (1989), pp. 553-558
[78.]
Morreale M., Tsirigotis M., Hughes M.D., Brumfitt W., McIntyre N., Burroughs A.K..
Significant bacteriuria has prognostic significance in primary biliary cirrhosis.
J Hepatol, 9 (1989), pp. 149-158
[79.]
Rosenstein I.J., Hazlehurst G.R., Burroughs A.K., Epstein O., Sherlock S., Brumfitt W..
Recurrent bacteriuria and primary biliary cirrhosis: ABO blood group, P1 blood group, and secretor status.
J Clin Pathol, 37 (1984), pp. 1055-1058
[80.]
Smyk D., Mytilinaiou M.G., Rigopoulou E.I., Bogdanos D.P..
PBC triggers in water reservoirs, coal mining areas and waste disposal sites: from Newcastle to New York.
Dis Markers, 29 (2010), pp. 337-344
[81.]
Smyk D., Rigopoulou E.I., Baum H., Burroughs A.K., Vergani D., Bogdanos D.P..
Autoimmunity and Environment: Am I at risk?.
Clin Rev Allergy Immunol, (2011),
[82.]
Corpechot C., Chretien Y., Chazouilleres O., Poupon R..
Demographic, lifestyle, medical and familial factors associated with primary biliary cirrhosis.
J Hepatol, 53 (2010), pp. 162-169
[83.]
Corpechot C., Gaouar F., Chretien Y., Johanet C., Chazouilleres O., Poupon R..
Smoking as an Independent Risk Factor of Liver Fibrosis in Primary Biliary Cirrhosis.
[84.]
Gershwin M.E., Selmi C., Worman H.J., Gold E.B., Watnik M., Utts J., et al.
Risk factors and comorbidities in primary biliary cirrhosis: a controlled interview-based study of 1032 patients.
Hepatology, 42 (2005), pp. 1194-1202
[85.]
Parikh-Patel A., Gold E.B., Worman H., Krivy K.E., Gershwin M.E..
Risk factors for primary biliary cirrhosis in a cohort of patients from the United States.
Hepatology, 33 (2001), pp. 16-21
[86.]
Prince M.I., Ducker S.J., James O.F..
Case-control studies of risk factors for primary biliary cirrhosis in two United Kingdom populations.
[87.]
Liang Y., Yang Z., Zhong R..
Smoking, family history and urinary tract infection are associated with primary biliary cirrhosis: A meta-analysis.
Hepatol Res, 41 (2011), pp. 572-578
[88.]
Berta L., Frairia R., Fortunati N., Fazzari A., Gaidano G..
Smoking effects on the hormonal balance of fertile women.
Horm Res, 37 (1992), pp. 45-48
[89.]
Bijl M., Horst G., Limburg P.C., Kallenberg C.G..
Effects of smoking on activation markers, Fas expression and apoptosis of peripheral blood lymphocytes.
Eur J Clin Invest, 31 (2001), pp. 550-553
[90.]
Churg A., Zay K., Shay S., Xie C., Shapiro S.D., Hendricks R., et al.
Acute cigarette smoke-induced connective tissue breakdown requires both neutrophils and macrophage metalloelastase in mice.
Am J Respir Cell Mol Biol, 27 (2002), pp. 368-374
[91.]
Cozen W., Diaz-Sanchez D., James Gauderman W., Zadnick J., Cockburn M.G., Gill P.S., et al.
Th1 and Th2 cytokines and IgE levels in identical twins with varying levels of cigarette consumption.
J Clin Immunol, 24 (2004), pp. 617-622
[92.]
Dev A., Patel K., Conrad A., Blatt L.M., McHutchison J.G..
Relationship of smoking and fibrosis in patients with chronic hepatitis C.
Clin Gastroenterol Hepatol, 4 (2006), pp. 797-801
[93.]
Kannel W.B., D’Agostino R.B., Belanger A.J..
Fibrinogen, cigarette smoking, and risk of cardiovascular disease: insights from the Framingham Study.
Am Heart J, 113 (1987), pp. 1006-1010
[94.]
Petitti D.B., Kipp H..
The leukocyte count: associations with intensity of smoking and persistence of effect after quitting.
Am J Epidemiol, 123 (1986), pp. 89-95
[95.]
Reiman R.M., Thompson R.W., Feng C.G., Hari D., Knight R., Cheever A.W., et al.
Interleukin-5 (IL-5) augments the progression of liver fibrosis by regulating IL-13 activity.
Infect Immun, 74 (2006), pp. 1471-1479
[96.]
Seagrave J., Barr E.B., March T.H., Nikula K.J..
Effects of cigarette smoke exposure and cessation on inflammatory cells and matrix metalloproteinase activity in mice.
Exp Lung Res, 30 (2004), pp. 1-15
[97.]
Soldin O.P., Makambi K.H., Soldin S.J., O’Mara D.M..
Steroid hormone levels associated with passive and active smoking.
Steroids, (2011),
[98.]
Sugimoto R., Enjoji M., Nakamuta M., Ohta S., Kohjima M., Fukushima M., et al.
Effect of IL-4 and IL-13 on collagen production in cultured LI90 human hepatic stellate cells.
[99.]
Tsochatzis E., Papatheodoridis G.V., Manolakopoulos S., Tiniakos D.G., Manesis E.K., Archimandritis A.J..
Smoking is associated with steatosis and severe fibrosis in chronic hepatitis C but not B.
Scand J Gastroenterol, 44 (2009), pp. 752-759
[100.]
Whetzel C.A., Corwin E.J., Klein L.C..
Disruption in Th1/Th2 immune response in young adult smokers.
Addict Behav, 32 (2007), pp. 1-8
[101.]
Zein C.O..
Clearing the smoke in chronic liver diseases.
Hepatology, 51 (2010), pp. 1487-1490
[102.]
Howel D., Fischbacher C.M., Bhopal R.S., Gray J., Metcalf J.V., James O.F..
An exploratory population-based case-control study of primary biliary cirrhosis.
Hepatology, 31 (2000), pp. 1055-1060
[103.]
Bataller R..
Time to ban smoking in patients with chronic liver diseases.
Hepatology, 44 (2006), pp. 1394-1396
[104.]
Corrao G., Lepore A.R., Torchio P., Valenti M., Galatola G., D’Amicis A., et al.
The effect of drinking coffee and smoking cigarettes on the risk of cirrhosis associated with alcohol consumption. A case-control study. Provincial Group for the Study of Chronic Liver Disease.
Eur J Epidemiol, 10 (1994), pp. 657-664
[105.]
El-Zayadi A.R..
Heavy smoking and liver.
World J Gastroenterol, 12 (2006), pp. 6098-6101
[106.]
Hezode C., Lonjon I., Roudot-Thoraval F., Mavier J.P., Pawlotsky J.M., Zafrani E.S., et al.
Impact of smoking on histological liver lesions in chronic hepatitis C.
Gut, 52 (2003), pp. 126-129
[107.]
Keller A.Z..
Cirrhosis of the liver, alcoholism and heavy smoking associated with cancer of the mouth and pharynx.
Cancer, 20 (1967), pp. 1015-1022
[108.]
Klatsky A.L., Armstrong M.A..
Alcohol, smoking, coffee, and cirrhosis.
Am J Epidemiol, 136 (1992), pp. 1248-1257
[109.]
Zein C.O., Beatty K., Post A.B., Logan L., Debanne S., McCullough A.J..
Smoking and increased severity of hepatic fibrosis in primary biliary cirrhosis: A cross validated retrospective assessment.
Hepatology, 44 (2006), pp. 1564-1571
[110.]
Kaviratne M., Hesse M., Leusink M., Cheever A.W., Davies S.J., McKerrow J.H., et al.
IL-13 activates a mechanism of tissue fibrosis that is completely TGF-beta independent.
J Immunol, 173 (2004), pp. 4020-4029
[111.]
Pessione F., Ramond M.J., Njapoum C., Duchatelle V., Degott C., Erlinger S., et al.
Cigarette smoking and hepatic lesions in patients with chronic hepatitis C.
Hepatology, 34 (2001), pp. 121-125
[112.]
Costenbader K.H., Karlson E.W..
Cigarette smoking and autoimmune disease: what can we learn from epidemiology?.
Lupus, 15 (2006), pp. 737-745
[113.]
Sopori M..
Effects of cigarette smoke on the immune system.
Nat Rev Immunol, 2 (2002), pp. 372-377
[114.]
Shan M., Cheng H.F., Song L.Z., Roberts L., Green L., HackenBitar J., et al.
Lung myeloid dendritic cells coordinately induce TH1 and TH17 responses in human emphysema.
Sci Transl Med, 1 (2009), pp. 4-10
[115.]
Stefanska A.M., Walsh P.T..
Chronic obstructive pulmonary disease: evidence for an autoimmune component.
Cell Mol Immunol, 6 (2009), pp. 81-86
[116.]
Tsoumakidou M., Tsiligianni I., Tzanakis N..
Mechanisms of altered cell immunity and cytotoxicity in COPD.
Curr Drug Targets, 12 (2011), pp. 450-459
[117.]
Majori M., Corradi M., Caminati A., Cacciani G., Bertacco S., Pesci A..
Predominant TH1 cytokine pattern in peripheral blood from subjects with chronic obstructive pulmonary disease.
J Allergy Clin Immunol, 103 (1999), pp. 458-462
[118.]
Harada K., Van de Water J., Leung P.S., Coppel R.L., Ansari A., Nakanuma Y., et al.
In situ nucleic acid hybridization of cytokines in primary biliary cirrhosis: predominance of the Th1 subset.
Hepatology, 25 (1997), pp. 791-796
[119.]
Ala A., Stanca C.M., Bu-Ghanim M., Ahmado I., Branch A.D., Schiano T.D., et al.
Increased prevalence of primary biliary cirrhosis near Superfund toxic waste sites.
Hepatology, 43 (2006), pp. 525-531
Copyright © 2012. Fundación Clínica Médica Sur, A.C.
Article options
es en pt

¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

Are you a health professional able to prescribe or dispense drugs?

Você é um profissional de saúde habilitado a prescrever ou dispensar medicamentos