Skip to main content

Migration of microsomal autoantigens to the hepatocyte plasma membrane

  • Chapter
Immunology and Liver

Part of the book series: Falk Symposium ((FASS,volume 114))

  • 122 Accesses

Abstract

Autoantibodies against microsomal autoantigens are present in some forms of drug-induced hepatitis, alcohol-induced liver injury, idiopathic autoimmune hepatitis, and viral hepatitis. For these autoantibodies to play a role in immune destruction the microsomal autoantigens would have to be inserted in, or transported to, the plasma membrane (PM) and would have to be exposed, at least in part, on the outer surface of the PM. The present review is mainly concerned with the immunogenicity, cellular transport, and PM expression of cytochromes P450 (CYP). Other microsomal autoantigens are briefly discussed.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 129.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 169.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 169.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Preview

Unable to display preview. Download preview PDF.

Unable to display preview. Download preview PDF.

References

  1. Gonzales FJ, Nebert DW. Evolution of the cytochrome P-450 gene superfamily: animal-plant ‘warfare’, molecular drive and human genetic differences in drug oxidation. Trends Genet. 1990;6:182–186.

    Article  Google Scholar 

  2. Germain RN. MHC-dependent antigen processing and peptide presentation: providing ligands for T lymphocyte activation. Cell. 1994;76:287–299.

    Article  PubMed  CAS  Google Scholar 

  3. Zimmerman HJ, Lewis JH, Ishak KG, Maddrey W. Ticrynafen-associated hepatic injury: analysis of 340 cases. Hepatology. 1984;4:315–323.

    Article  PubMed  CAS  Google Scholar 

  4. Lopez Garcia MP, Dansette PM, Valadon P et al. Human-liver cytochromes P-450 expressed in yeast as tools for reactive-metabolite formation studies. Oxidative activation of tienilic acid by cytochromes P-450 2C9 and 2C10. Eur J Biochem. 1993;213:223–232.

    Article  Google Scholar 

  5. Lecoeur S, Bonierbale E, Challin D et al. Specificity of in vitrocovalent binding of tienilic acid metabolites to human liver microsomes in relationship with the type of hepatotoxicity: comparison with two directly hepatotoxic drugs. Chem Res Toxicol. 1994;7:434–442.

    Article  PubMed  CAS  Google Scholar 

  6. Lopez-Garcia MP, Dansette P, Mansuy D. Thiophene derivatives as new mechanism-based inhibitors of cytochrome P450: inactivation of yeast-expressed human liver P450 2C9 by tienilic acid. Biochemistry. 1994;33:166–175.

    Article  PubMed  CAS  Google Scholar 

  7. Homberg JC, André C, Abuaf N. A new anti-liver-kidney-microsome antibody (anti-LKM2) in tienilic acid-induced hepatitis. Clin Exp Immunol. 1984;55:561–570.

    PubMed  CAS  Google Scholar 

  8. Beaune PH, Dansette PM, Mansuy D et al. Human anti-endoplasmic reticulum auto-antibodies appearing in a drug induced hepatitis are directed against a human liver cytochrome P-450 that hydroxylates the drug. Proc Natl Acad Sci USA. 1987;84:551–555.

    Article  PubMed  CAS  Google Scholar 

  9. Lecoeur S, André C, Beaune PH. Tienilic acid-induced autoimmune hepatitis: anti-liver and-kidney microsomal type 2 autoantibodies recognize a three-site conformational epitope on cytochrome P4502C9. Mol Pharmacol. 1996;50:326–333.

    PubMed  CAS  Google Scholar 

  10. Pons C, Dansette PM, Amar C et al. Detection of human hepatitis anti-liver kidney microsomes (LKM2) autoantibodies on rat liver sections is predominantly due to reactivity with rat liver P-450 IIC11. J Pharmacol Exp Ther 1991;259:1328–1334.

    PubMed  CAS  Google Scholar 

  11. Bourdi M, Larrey D, Nataf J et al. Anti-liver endoplasmic reticulum autoantibodies are directed against human cytochrome P-450 IA2: a specific marker of dihydralazine-induced hepatitis. J Clin Invest. 1990;85:1967–1973.

    Article  PubMed  CAS  Google Scholar 

  12. Bourdi M, Tinel M, Beaune P, Pessayre D. Interactions of dihydralazine with cytochromes P4501A: a possible explanation for the appearance of anti-P4501A2 autoantibodies. Mol Pharmacol. 1994;45:1287–1295.

    PubMed  CAS  Google Scholar 

  13. Spracklin DK, Hankins DC, Fisher JM, Thummel KE, Kharasch ED. Cytochrome P4502E1 is the principal catalyst of human oxidative halothane metabolism in vitro. J Pharmacol Exp Ther. 1997;281:400–411.

    PubMed  CAS  Google Scholar 

  14. Eliasson E, Kenna JG. Cytochrome P4502E1 is a cell surface autoantigen in halothane hepatitis. Mol Pharmacol 1996;50:573–582.

    PubMed  CAS  Google Scholar 

  15. Bourdi M, Chen W, Peter RM et al. Human cytochrome P4502E1 is a major autoantigen associated with halothane hepatitis. Chem Res Toxicol. 1996; 9:1159–1166.

    Article  PubMed  CAS  Google Scholar 

  16. Salmela KS, Kessova IG, Tsyrlov IB, Lieber CS. Respective roles of human cytochrome P-4502E1, 1A2, and 3A4 in the hepatic microsomal ethanol oxidizing system. Alcohol: Clin Exp Res. 1998;22:2125–2132.

    Article  CAS  Google Scholar 

  17. Behrens UJ, Hoerner M, Lasker JM, Lieber CS. Formation of acetaldehyde adducts with ethanol-inducible P450IIE1 in vivo. Biochem Biophys Res Commun. 1988;154:584–590.

    Article  PubMed  CAS  Google Scholar 

  18. Albano E, Tomasi A, Goria-Gatti L, Dianzani MU. Spin trapping of free radical species produced during the microsomal metabolism of ethanol. Chem Biol Interact. 1988;65:233–234.

    Article  Google Scholar 

  19. Clot P, Albano E, Eliasson E et al. Cytochrome P4502E1 hydroxy ethyl radical adducts as the major antigen in autoantibody formation among alcoholics. Gastroenterology. 1996; 111:206–216.

    Article  PubMed  CAS  Google Scholar 

  20. Lytton SD, Helander A, Zhang-Gouillon ZQ et al. Autoantibodies against cytochromes P-4502E1 and P-4503A in alcoholics. Mol Pharmacol. 1999;55:223–233.

    PubMed  CAS  Google Scholar 

  21. Leeder JS, Riley RJ, Cook VA, Spielberg SP. Human anti-cytochrome P450 antibodies in aromatic anticonvulsant-induced hypersensitivity reactions. J Pharmacol Exp Ther. 1992; 263:360–367.

    PubMed  CAS  Google Scholar 

  22. Riley RJ, Smith G, Wolf CR, Cook VA, Leeder JS. Human anti-endoplasmic reticulum autoantibodies produced in aromatic anticonvulsant hypersensitivity reactions recognize rodent CYP3A proteins and a similarly regulated human P450 enzyme(s). Biochem Biophys Res Commun. 1993;191:32–40.

    Article  PubMed  CAS  Google Scholar 

  23. Leeder JS, Lu X, Timsit Y, Gaedigk A. Non-monooxygenase cytochromes P450 as potential human autoantigens in anticonvulsant hypersensitivity reactions. Pharmacogenetics. 1998;8:211–225.

    Article  PubMed  CAS  Google Scholar 

  24. Beaune PH, Pessayre D, Dansette PM, Mansuy D, Manns M. Autoantibodies against cytochromes P450: role in human diseases. Adv Pharmacol. 1994;30:199–245.

    Article  PubMed  CAS  Google Scholar 

  25. Robin MA, Maratrat M et al. Antigenic targets in tienilic acid hepatitis. Both cytochrome P450 2C11 and 2C11-tienilic acid adducts are transported to the plasma membrane of rat hepatocytes and recognized by human sera. J Clin Invest 1996;98:1471–1480.

    Article  PubMed  CAS  Google Scholar 

  26. Bonierbale E. Mécanisme des hepatites immunoallergiques induites par l’acide tiénilique: activation métabolique de ce médicament et fixation covalente sur les cytochromes P4502C chez l’homme et le rat. Thèse de doctorat de l’Université Paris V, 1996.

    Google Scholar 

  27. Clot P, Parola M, Bellomo G et al. Plasma membrane hydroxyethyl radical adducts cause antibody-dependent cytotoxicity in rat hepatocytes exposed to alcohol. Gastroenterology. 1997;113:265–276.

    Article  PubMed  CAS  Google Scholar 

  28. Robin MA, Le Roy M, Descatoire V, Pessayre D. Plasma membrane cytochromes P450 as neoantigens and autoimmune targets in drug-induced hepatitis. J Hepatol. 1997;26(suppl.l):23–30.

    Article  PubMed  CAS  Google Scholar 

  29. Pessayre D, Larrey D, Biour M. Drug-induced liver injury. In: Bircher J, Benhamon JP, Mclntyre N, Rizalto M, Rodes J, editors. Oxford Textbook of Clinical Hepatology 2nd edn. Oxford: Oxford University Press; 1999:1260–1315.

    Google Scholar 

  30. Guilbert B, Dighiero G, Avrameas S. Naturally occurring antibodies against nine common antigens in normal humans. I. Detection, isolation and characterization. J Immunol. 1982; 28:2779–2783.

    Google Scholar 

  31. Parker DC. T-cell dependent B-cell activation. Annu Rev Immunol. 1993;11:331–360.

    Article  PubMed  CAS  Google Scholar 

  32. Cambier JC, Morrisson DC, Chien MM, Lehmann KR. Modeling of T-cell contact-dependent B cell activation. IL-4 and antigen receptor ligation primes quiescent B cells to mobilize calcium in response to la cross-linking. J Immunol. 1991;146:2075–2082.

    PubMed  CAS  Google Scholar 

  33. Pessayre D. Role of reactive metabolites in drug-induced hepatitis. J Hepatol. 1995; 23(suppl.1): 16–24.

    PubMed  CAS  Google Scholar 

  34. Gut J, Christen U, Huwyler J. Mechanism of halothane toxicity: novel insights. Pharmacol Ther. 1993;58:133–155.

    Article  PubMed  CAS  Google Scholar 

  35. Berson A, Fréneaux D, Larrey D et al Possible role of HLA in hepatotoxicity. An exploratory study with drug-induced idiosyncratic hepatitis. J Hepatol. 1994;20:336–342.

    Article  PubMed  CAS  Google Scholar 

  36. Homberg JC, Abuaf N, Bernard O et al Chronic active hepatitis associated with anti-liver/kidney microsome antibody type 1: a second type of ‘autoimmune’ hepatitis. Hepatology. 1987;7:1333–1339.

    Article  PubMed  CAS  Google Scholar 

  37. Zanger UM, Hauri HP, Loeper J, Homberg JC, Meyer UA. Antibodies against human cytochrome P-450dbl in autoimmune hepatitis type II. Proc Natl Acad Sci USA. 1988;85:8256–8260.

    Article  PubMed  CAS  Google Scholar 

  38. Manns MP, Johnson EF, Griffin KJ, Tan EM, SulliVan KF. Major antigen of liver kidney microsomal autoantibodies in idiopathic autoimmune hepatitis is cytochrome P450dbl. J Clin Invest. 1989;83:1066–1072.

    Article  PubMed  CAS  Google Scholar 

  39. Manns MP, Griffin KJ, SulliVan KF, Johnson EF. LKM-1 autoantibodies recognize a short linear sequence in P450IID6, a cytochrome P450 monooxygenase. J Clin Invest. 1991;88:1370–1378.

    Article  PubMed  CAS  Google Scholar 

  40. Duclos-Vallée JC, Hajoui O, Yamamoto AM, Jacqz-Aigrain E, Alvarez F. Conformational epitopes on CYP2D6 are recognized by liver/kidney microsomal antibodies. Gastroenterology. 1995;108:470–476.

    Article  PubMed  Google Scholar 

  41. Seeling R, Renz M, Bünger G, Schröter H, Seeling HP. Anti-LKM-1 antibodies determined by use of recombinant P4502D6 in ELISA and Western blot and their association with anti-HCV and HCV-RNA. Clin Exp Immunol. 1993;92:373–380.

    Article  Google Scholar 

  42. Dalekos GN, Wedemeyer H, Obermayer-Sraub P et al Epitope mapping of cytochrome P450 2D6 autoantigen in patients with chronic hepatitis C under interferon-α treatment. J Hepatol. 1999 (In press).

    Google Scholar 

  43. Ahonen P, Myllärmiemi S, Sipilä I, Perheentupa J. Clinical variation of autoimmune polyen-docrinopathy-candidiasis-ectodermal dystrophy (APECED) in a series of 68 patients. N Engl J Med. 1990;322:1829–1836.

    Article  PubMed  CAS  Google Scholar 

  44. Clemente MG, Meloni A, Obermayer-Straub P, Frau F, Manns MP, De Virgiliis S. Two cytochromes P450 are major hepatocellular autoantigens in autoimmune polyglandular syndrome type 1. Gastroenterology. 1998;114:324–328.

    Article  PubMed  CAS  Google Scholar 

  45. DeLemos-Chiarandini C, Alvarez F, Bernard O, Homberg JC, Kreibich G. Anti-liver microsome antibody is a marker for the rat hepatocyte endoplasmic reticulum. Hepatology. 1987;7:468–475.

    Article  PubMed  Google Scholar 

  46. Trautwein C, Gerken G, Löhr H, Meyer Zum Büschenfelde KH, Manns M. Lack of surface expression for the B-cell autoepitope of cytochrome P450IID6 evidenced by flow cytometry. Z Gastroenterol. 1993;31:225–230.

    PubMed  CAS  Google Scholar 

  47. Yamamoto AM, Mura C, DeLemos-Chiarandini C, Krishnamoorthy R, Alvarez F. Cytochrome P-450IID6 recognized by LKM1 antibody is not exposed on the surface of hepatocytes. Clin Exp Immunol. 1993;92:381–390.

    Article  PubMed  CAS  Google Scholar 

  48. Jarasch ED, Kartenberg J, Bruder G, Fink A, Morré DJ, Franke WW. B-type cytochromes in plasma membranes isolated from rat liver, in comparison with those of endomembranes. J Cell Biol. 1979;80:37–52.

    Article  PubMed  CAS  Google Scholar 

  49. Stasiecki P, Oesch F. Distribution of enzymes involved in metabolic activation of polycyclic aromatic hydrocarbons among rat liver endomembranes and plasma membranes. Eur J Cell Biol. 1980;21:79–92.

    PubMed  CAS  Google Scholar 

  50. Lenzi M, Bianchi FB, Cassani F, Pisi E. Liver cell surface expression of the antigen reacting with liver-kidney microsomal antibody (LKM). Clin Exp Immunol. 1984;55:36–40.

    PubMed  CAS  Google Scholar 

  51. Satoh H, Davies HW, Takemura T, Gillette JR, Maeda K, Pohl LR. An immunochemical approach to investigate the mechanism of halothane-induced hepatitis. Progr Drug Metab. 1987;10:187–205.

    CAS  Google Scholar 

  52. Loeper J, Descatoire V, Maurice M et al. Presence of functional cytochrome P-450 on isolated rat hepatocyte plasma membrane. Hepatology. 1990;11:850–858.

    Article  PubMed  CAS  Google Scholar 

  53. Wu D, Cederbaum AI. Presence of functionally active cytochrome P-450 IIE1 in the plasma membrane of rat hepatocytes. Hepatology. 1992;15:515–524.

    Article  PubMed  CAS  Google Scholar 

  54. Loeper J, Descatoire V, Maurice M et al. Cytochromes P-450 in human hepatocyte plasma membrane: recognition by several autoantibodies. Gastroenterology. 1993;104:203–216.

    PubMed  CAS  Google Scholar 

  55. Sakaguchi M, Mihara K, Sato R. A short amino-terminal segment of microsomal cytochrome P-450 functions both as an insertion signal and a stop-transfer sequence. EMBO J. 1987;6:2425–2431.

    PubMed  CAS  Google Scholar 

  56. Neve EPA, Eliasson E, Pronzato MA, Albano E, Marinari U, Ingelman-Sundberg M. Enzyme-specific transport of rat liver cytochrome P450 to the Golgi apparatus. Arch Biochem Biophys. 1996;333:459–465.

    Article  PubMed  CAS  Google Scholar 

  57. Robin MA, Maratrat M, Loeper J et al. Cytochrome P4502B follows a vesicular route to the plasma membrane in cultured rat hepatocytes. Gastroenterology. 1995;108:1110–1123.

    Article  PubMed  CAS  Google Scholar 

  58. Loeper J, Louérat-Oriou B, Duport C, Pompon D. Yeast expressed cytochrome P450 2D6 (CYP2D6) exposed on the external face of plasma membrane is functionally competent. Mol Pharmacol. 1998;54:8–13.

    PubMed  CAS  Google Scholar 

  59. De Lemos-Chiarandini C, Frey AB, Sabatini DD, Kreibich G. Determination of the membrane topology of the phenobarbital-inducible rat liver cytochrome P-450 isoenzyme PB-4 using site-specific antibodies. J Cell Biol. 1987;104:209–219.

    Article  PubMed  Google Scholar 

  60. Vergères G, Winterhalter KH, Richter C. Localization of the N-terminal methionine of rat liver cytochrome P-450 in the lumen of the endoplasmic reticulum. Biochim Biophys Acta. 1991;1063:235–241.

    Article  PubMed  Google Scholar 

  61. Black SD, Martin ST, Smith CA. Membrane topology of liver microsomal cytochrome P450 2B4 determined via monoclonal antibodies directed to the halt transfer signal. Biochemistry. 1994;33:6945–6951.

    Article  PubMed  CAS  Google Scholar 

  62. Kida Y, Ohgiya S, Mihara K, Sakaguchi M. Membrane topology of NADPH-cytochrome P450 reductase on the endoplasmic reticulum. Arch Biochem Biophys. 1998;351:175–179.

    Article  PubMed  CAS  Google Scholar 

  63. Szczesna-Skorupa E, Ahn K, Chen CD, Doray B, Kemper B. The cytoplasmic and N-terminal transmembrane domains of cytochrome P450 contain independent signals for retention in the endoplasmic reticulum. J Biol Chem. 1995;41:24327–24332.

    Google Scholar 

  64. Loeper J, Le Berre A, Pompon D. Topology inversion of CYP2D6 in the endoplasmic reticulum is not required for plasma membrane transport. Mol Pharmacol. 1998;53:408–414.

    PubMed  CAS  Google Scholar 

  65. Addya S, Anandatheerthavarada HK, Biswas G, Bhagwat SV, Mullick J, Avadhani NG. Targeting of NH2-terminal-processed microsomal protein to mitochondria: a novel pathway for the biogenesis of hepatic mitochondrial P450MT2. J Cell Biol. 1997;139:589–599.

    Article  PubMed  CAS  Google Scholar 

  66. Zachowski A, Henry JP, Devaux PR Control of transmembrane lipid asymmetry in chromaffin granules by an ATP-dependent protein. Nature. 1989;340:75–76.

    Article  PubMed  CAS  Google Scholar 

  67. Neuberger J, Williams R. Immune mechanisms in tienilic acid associated hepatotoxicity. Gut. 1989;30:515–519.

    Article  PubMed  CAS  Google Scholar 

  68. Vergani D, Mieli-Vergani G, Alberti A et al. Antibodies to the surface of halothane-altered rabbit hepatocytes in patients with severe halothane-associated hepatitis. N Engl J Med. 1980;303:66–71.

    Article  PubMed  CAS  Google Scholar 

  69. Philipp T, Durazzo M, Trautwein C et al Recognition of uridine diphosphate glucuronosyl transferases by LKM-3 antibodies in chronic hepatitis D. Lancet. 1994;344:578–581.

    Article  PubMed  CAS  Google Scholar 

  70. Yokota H, Yuasa A, Sata R. Topological disposition of UDP glucuronosyltransferase in rat liver microsomes. J Biochem. 1992;112:192–196.

    PubMed  CAS  Google Scholar 

  71. Meech R, Mackenzie PI. Determinants of UDP glucuronosyltransferase membrane association and residency in the endoplasmic reticulum. Arch Biochem Biophys. 1998;356:77–85.

    Article  PubMed  CAS  Google Scholar 

  72. Alves C, Von Dippe P, Amoui M, Levy D. Bile acid transport into hepatocyte smooth endoplasmic reticulum vesicles is mediated by microsomal epoxide hydrolase, a membrane protein exhibiting two distinct topological orientations. J Biol Chem. 1993;268:20148–20155.

    PubMed  CAS  Google Scholar 

  73. Wesierska-Gadek J, Grimm R, Hitchman E, Penner E. Members of the glutathione S-trans-ferase gene family are antigens in autoimmune hepatitis. Gastroenterology. 1998;114:329–335.

    Article  PubMed  CAS  Google Scholar 

  74. Andersson C, Weinander R, Lundqvist G, DePierre JW, Morgenstern R. Functional and structural membrane topology of rat liver microsomal glutathione transferase. Biochim Biophys Acta. 1994;1204:298–304.

    Article  PubMed  CAS  Google Scholar 

  75. Luzio JP, Brake B, Banting G, Howell KE, Braghetta P, Stanley KK. Identification, sequencing and expression of an integral membrane protein of the trans-Golgi network. Biochem J. 1990;270:97–

    PubMed  CAS  Google Scholar 

  76. Horbach ME, Sies H, Akerboom TPM. Purification of rat liver plasma membrane glutathione transferase. Eur J Biochem. 1994;222:91–96.

    Article  PubMed  CAS  Google Scholar 

  77. Treichel U, Gerken G, Rossol S, Rotthauwe HW, Meyer zum Büschenfelde KH, Poralla T. Autoantibodies against the human asialoglycoprotein receptor: effects of therapy in autoimmune and virus-induced chronic active hepatitis. J Hepatol. 1993;19:55–63.

    Article  PubMed  CAS  Google Scholar 

  78. Spiess M. The asialoglycoprotein receptor: a model for endocytic transport receptors. Biochemistry. 1990;29:10009–10018.

    Article  PubMed  CAS  Google Scholar 

Download references

Authors

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2000 Springer Science+Business Media Dordrecht

About this chapter

Cite this chapter

Pessayre, D., Descatoire, V., Dansette, P., Loeper, J., Berson, A., Robin, MA. (2000). Migration of microsomal autoantigens to the hepatocyte plasma membrane. In: Manns, M.P., Paumgartner, G., Leuschner, U. (eds) Immunology and Liver. Falk Symposium, vol 114. Springer, Dordrecht. https://doi.org/10.1007/978-94-011-4000-3_3

Download citation

  • DOI: https://doi.org/10.1007/978-94-011-4000-3_3

  • Publisher Name: Springer, Dordrecht

  • Print ISBN: 978-94-010-5768-4

  • Online ISBN: 978-94-011-4000-3

  • eBook Packages: Springer Book Archive

Publish with us

Policies and ethics