Skip to main content
Top
Published in: Journal of Clinical Immunology 3/2008

01-05-2008

Interferon-α Induces Up-regulation and Nuclear Translocation of the Ro52 Autoantigen as Detected by a Panel of Novel Ro52-specific Monoclonal Antibodies

Authors: Linn Strandberg, Aurelie Ambrosi, Alexander Espinosa, Lars Ottosson, Maija-Leena Eloranta, Wei Zhou, Åse Elfving, Edward Greenfield, Vijay K. Kuchroo, Marie Wahren-Herlenius

Published in: Journal of Clinical Immunology | Issue 3/2008

Login to get access

Abstract

Interferon-α (IFN-α) has been implicated in the pathogenesis of Sjögren’s syndrome and systemic lupus erythematosus. Ro52, which was recently identified as an E3 ligase with anti-proliferative and pro-apoptotic properties, is a major autoantigen targeted in both these conditions. Microarray analyses have indicated up-regulation of Ro52 by INF-α, and the objective of the present study was to address the potential link between IFN-α and Ro52. To investigate the influence of IFN-α on Ro52 protein levels and cellular localization, we generated a panel of monoclonal antibodies to different domains of Ro52. These novel monoclonal antibodies were characterized by immunoprecipitation, Western blot, and enzyme-linked immunosorbent assay using cell lysates, recombinant Ro52 protein, and synthetic peptides. Ro52 was up-regulated in HeLa cells and human B cells at the messenger RNA and protein levels in response to IFN-α stimulation as detected by reverse transcriptase polymerase chain reaction and Western blot. After up-regulation, Ro52 translocated from the cytoplasm to the nucleus. The nuclear translocation of Ro52 was observed after staining with generated monoclonal antibodies specific for both the RING, coiled-coil, and B30.2 domains of Ro52 and the nuclear translocation of Ro52 preceded IFN-α-induced apoptotic cell death detected by caspase-3 and TUNEL staining in the treated cultures. In conclusion, our data show that IFN-α first induces up-regulation of Ro52 protein and then prompts translocation of the up-regulated Ro52 protein in to the nucleus. The translocation precedes apoptosis of the IFN-α exposed cells, suggesting a role for Ro52 in mediating the anti-proliferative or pro-apoptotic effects of the autoimmune-related cytokine IFN-α.
Literature
1.
go back to reference Kirkwood JM, Ernstoff MS. Interferons in the treatment of human cancer. J Clin Oncol 1984;2:336–52.PubMed Kirkwood JM, Ernstoff MS. Interferons in the treatment of human cancer. J Clin Oncol 1984;2:336–52.PubMed
2.
go back to reference Rönnblom LE, Alm GV, Oberg KE. Autoimmunity after alpha-interferon therapy for malignant carcinoid tumors. Ann Intern Med 1991;115:178–83.PubMed Rönnblom LE, Alm GV, Oberg KE. Autoimmunity after alpha-interferon therapy for malignant carcinoid tumors. Ann Intern Med 1991;115:178–83.PubMed
3.
go back to reference Bengtsson AA, Sturfelt G, Truedsson L, Blomberg J, Alm G, Vallin H, Ronnblom L. Activation of type I interferon system in systemic lupus erythematosus correlates with disease activity but not with antiretroviral antibodies. Lupus 2000;9:664–71.PubMedCrossRef Bengtsson AA, Sturfelt G, Truedsson L, Blomberg J, Alm G, Vallin H, Ronnblom L. Activation of type I interferon system in systemic lupus erythematosus correlates with disease activity but not with antiretroviral antibodies. Lupus 2000;9:664–71.PubMedCrossRef
4.
go back to reference Bennett L, Palucka AK, Arce E, Cantrell V, Borvak J, Banchereau J, Pascual V. Interferon and granulopoiesis signatures in systemic lupus erythematosus blood. J Exp Med 2003;197:711–23.PubMedCrossRef Bennett L, Palucka AK, Arce E, Cantrell V, Borvak J, Banchereau J, Pascual V. Interferon and granulopoiesis signatures in systemic lupus erythematosus blood. J Exp Med 2003;197:711–23.PubMedCrossRef
5.
go back to reference Baechler EC, Batliwalla FM, Karypis G, Gaffney PM, Ortmann WA, Espe KJ, Shark KB, Grande WJ, Hughes KM, Kapur V, Gregersen PK, Behrens TW. Interferon-inducible gene expression signature in peripheral blood cells of patients with severe lupus. Proc Natl Acad Sci U S A 2003;100:2610–5.PubMedCrossRef Baechler EC, Batliwalla FM, Karypis G, Gaffney PM, Ortmann WA, Espe KJ, Shark KB, Grande WJ, Hughes KM, Kapur V, Gregersen PK, Behrens TW. Interferon-inducible gene expression signature in peripheral blood cells of patients with severe lupus. Proc Natl Acad Sci U S A 2003;100:2610–5.PubMedCrossRef
6.
go back to reference Båve U, Nordmark G, Lövgren T, Rönnelid J, Cajander S, Eloranta ML, Alm GV, Rönnblom L. Activation of the type I interferon system in primary Sjogren’s syndrome: a possible etiopathogenic mechanism. Arthritis Rheum 2005;52:1185–95.PubMedCrossRef Båve U, Nordmark G, Lövgren T, Rönnelid J, Cajander S, Eloranta ML, Alm GV, Rönnblom L. Activation of the type I interferon system in primary Sjogren’s syndrome: a possible etiopathogenic mechanism. Arthritis Rheum 2005;52:1185–95.PubMedCrossRef
7.
go back to reference Gottenberg JE, Cagnard N, Lucchesi C, Letourneur F, Mistou S, Lazure T, Jacques S, Ba N, Ittah M, Lepajolec C, Labetoulle M, Ardizzone M, Sibilia J, Fournier C, Chiocchia G, Mariette X. Activation of IFN pathways and plasmacytoid dendritic cell recruitment in target organs of primary Sjogren’s syndrome. Proc Natl Acad Sci U S A 2006;103:2770–5.PubMedCrossRef Gottenberg JE, Cagnard N, Lucchesi C, Letourneur F, Mistou S, Lazure T, Jacques S, Ba N, Ittah M, Lepajolec C, Labetoulle M, Ardizzone M, Sibilia J, Fournier C, Chiocchia G, Mariette X. Activation of IFN pathways and plasmacytoid dendritic cell recruitment in target organs of primary Sjogren’s syndrome. Proc Natl Acad Sci U S A 2006;103:2770–5.PubMedCrossRef
8.
go back to reference Der SD, Zhou A, Williams BR, Silverman RH. Identification of genes differentially regulated by interferon alpha, beta, or gamma using oligonucleotide arrays. Proc Natl Acad Sci U S A 1998;95:15623–8.PubMedCrossRef Der SD, Zhou A, Williams BR, Silverman RH. Identification of genes differentially regulated by interferon alpha, beta, or gamma using oligonucleotide arrays. Proc Natl Acad Sci U S A 1998;95:15623–8.PubMedCrossRef
9.
go back to reference de Veer MJ, Holko M, Frevel M, Walker E, Der S, Paranjape JM, Silverman RH, Williams BR. Functional classification of interferon-stimulated genes identified using microarrays. J Leukoc Biol 2001;69:912–20.PubMed de Veer MJ, Holko M, Frevel M, Walker E, Der S, Paranjape JM, Silverman RH, Williams BR. Functional classification of interferon-stimulated genes identified using microarrays. J Leukoc Biol 2001;69:912–20.PubMed
10.
go back to reference Pfeffer LM, Kim JG, Pfeffer SR, Carrigan DJ, Baker DP, Wei L, Homayouni R. Role of nuclear factor-kappaB in the antiviral action of interferon and interferon-regulated gene expression. J Biol Chem 2004;279:31304–11.PubMedCrossRef Pfeffer LM, Kim JG, Pfeffer SR, Carrigan DJ, Baker DP, Wei L, Homayouni R. Role of nuclear factor-kappaB in the antiviral action of interferon and interferon-regulated gene expression. J Biol Chem 2004;279:31304–11.PubMedCrossRef
11.
go back to reference Reymond A, Meroni G, Fantozzi A, Merla G, Cairo S, Luzi L, Riganelli D, Zanaria E, Messali S, Cainarca S, Guffanti A, Minucci S, Pelicci PG, Ballabio A. The tripartite motif family identifies cell compartments. Embo J 2001;20:2140–51.PubMedCrossRef Reymond A, Meroni G, Fantozzi A, Merla G, Cairo S, Luzi L, Riganelli D, Zanaria E, Messali S, Cainarca S, Guffanti A, Minucci S, Pelicci PG, Ballabio A. The tripartite motif family identifies cell compartments. Embo J 2001;20:2140–51.PubMedCrossRef
12.
go back to reference Espinosa A, Zhou W, Ek M, Hedlund M, Brauner S, Popovic K, Horvath L, Wallerskog T, Oukka M, Nyberg F, Kuchroo VK, Wahren-Herlenius M. The Sjogren’s syndrome-associated autoantigen Ro52 is an E3 ligase that regulates proliferation and cell death. J Immunol 2006;176:6277–85.PubMed Espinosa A, Zhou W, Ek M, Hedlund M, Brauner S, Popovic K, Horvath L, Wallerskog T, Oukka M, Nyberg F, Kuchroo VK, Wahren-Herlenius M. The Sjogren’s syndrome-associated autoantigen Ro52 is an E3 ligase that regulates proliferation and cell death. J Immunol 2006;176:6277–85.PubMed
13.
go back to reference Wada K, Kamitani T. Autoantigen Ro52 is an E3 ubiquitin ligase. Biochem Biophys Res Commun 2006;339:415–21.PubMedCrossRef Wada K, Kamitani T. Autoantigen Ro52 is an E3 ubiquitin ligase. Biochem Biophys Res Commun 2006;339:415–21.PubMedCrossRef
14.
go back to reference Sabile A, Meyer AM, Wirbelauer C, Hess D, Kogel U, Scheffner M, Krek W. Regulation of p27 degradation and S-phase progression by Ro52 RING finger protein. Mol Cell Biol 2006;26:5994–6004.PubMedCrossRef Sabile A, Meyer AM, Wirbelauer C, Hess D, Kogel U, Scheffner M, Krek W. Regulation of p27 degradation and S-phase progression by Ro52 RING finger protein. Mol Cell Biol 2006;26:5994–6004.PubMedCrossRef
15.
go back to reference Blange I, Ringertz NR, Pettersson I. Identification of antigenic regions of the human 52kD Ro/SS-A protein recognized by patient sera. J Autoimmun 1994;7:263–74.PubMedCrossRef Blange I, Ringertz NR, Pettersson I. Identification of antigenic regions of the human 52kD Ro/SS-A protein recognized by patient sera. J Autoimmun 1994;7:263–74.PubMedCrossRef
16.
go back to reference Ottosson L, Salomonsson S, Hennig J, Sonesson SE, Dorner T, Raats J, Kuchroo VK, Sunnerhagen M, Wahren-Herlenius M. Structurally derived mutations define congenital heart block-related epitopes within the 200-239 amino acid stretch of the Ro52 protein. Scand J Immunol 2005;61:109–18.PubMedCrossRef Ottosson L, Salomonsson S, Hennig J, Sonesson SE, Dorner T, Raats J, Kuchroo VK, Sunnerhagen M, Wahren-Herlenius M. Structurally derived mutations define congenital heart block-related epitopes within the 200-239 amino acid stretch of the Ro52 protein. Scand J Immunol 2005;61:109–18.PubMedCrossRef
17.
go back to reference Salomonsson S, Dorner T, Theander E, Bremme K, Larsson P, Wahren-Herlenius M. A serologic marker for fetal risk of congenital heart block. Arthritis Rheum 2002;46:1233–41.PubMedCrossRef Salomonsson S, Dorner T, Theander E, Bremme K, Larsson P, Wahren-Herlenius M. A serologic marker for fetal risk of congenital heart block. Arthritis Rheum 2002;46:1233–41.PubMedCrossRef
18.
go back to reference Fritsch C, Hoebeke J, Dali H, Ricchiuti V, Isenberg DA, Meyer O, Muller S. 52-kDa Ro/SSA epitopes preferentially recognized by antibodies from mothers of children with neonatal lupus and congenital heart block. Arthritis Res Ther 2005;8:R4.CrossRef Fritsch C, Hoebeke J, Dali H, Ricchiuti V, Isenberg DA, Meyer O, Muller S. 52-kDa Ro/SSA epitopes preferentially recognized by antibodies from mothers of children with neonatal lupus and congenital heart block. Arthritis Res Ther 2005;8:R4.CrossRef
19.
go back to reference Salomonsson S, Sonesson SE, Ottosson L, Muhallab S, Olsson T, Sunnerhagen M, Kuchroo VK, Thoren P, Herlenius E, Wahren-Herlenius M. Ro/SSA autoantibodies directly bind cardiomyocytes, disturb calcium homeostasis, and mediate congenital heart block. J Exp Med 2005;201:11–7.PubMedCrossRef Salomonsson S, Sonesson SE, Ottosson L, Muhallab S, Olsson T, Sunnerhagen M, Kuchroo VK, Thoren P, Herlenius E, Wahren-Herlenius M. Ro/SSA autoantibodies directly bind cardiomyocytes, disturb calcium homeostasis, and mediate congenital heart block. J Exp Med 2005;201:11–7.PubMedCrossRef
20.
go back to reference Sonesson SE, Salomonsson S, Jacobsson LA, Bremme K, Wahren-Herlenius M. Signs of first-degree heart block occur in one-third of fetuses of pregnant women with anti-SSA/Ro 52-kd antibodies. Arthritis Rheum 2004;50:1253–61.PubMedCrossRef Sonesson SE, Salomonsson S, Jacobsson LA, Bremme K, Wahren-Herlenius M. Signs of first-degree heart block occur in one-third of fetuses of pregnant women with anti-SSA/Ro 52-kd antibodies. Arthritis Rheum 2004;50:1253–61.PubMedCrossRef
21.
go back to reference Pourmand N, Pettersson I. The Zn2+ binding domain of the human Ro 52kD protein is a target for conformation-dependent autoantibodies. J Autoimmun 1998;11:11–7.PubMedCrossRef Pourmand N, Pettersson I. The Zn2+ binding domain of the human Ro 52kD protein is a target for conformation-dependent autoantibodies. J Autoimmun 1998;11:11–7.PubMedCrossRef
22.
go back to reference Eftekhari P, Salle L, Lezoualc, h F, Mialet J, Gastineau M, Briand JP, Isenberg DA, Fournie GJ, Argibay J, Fischmeister R, Muller S, Hoebeke J. Anti-SSA/Ro52 autoantibodies blocking the cardiac 5-HT4 serotoninergic receptor could explain neonatal lupus congenital heart block. Eur J Immunol 2000;30:2782–90.PubMedCrossRef Eftekhari P, Salle L, Lezoualc, h F, Mialet J, Gastineau M, Briand JP, Isenberg DA, Fournie GJ, Argibay J, Fischmeister R, Muller S, Hoebeke J. Anti-SSA/Ro52 autoantibodies blocking the cardiac 5-HT4 serotoninergic receptor could explain neonatal lupus congenital heart block. Eur J Immunol 2000;30:2782–90.PubMedCrossRef
23.
go back to reference Elagib KEE, Tengnér P, Levi M, Jonsson R, Thompson K, Natvig JB, Wahren-Herlenius M. Immunoglobulin variable genes and epitope recognition of human monoclonal anti-Ro 52-kd in primary Sjögren’s syndrome. Arthritis Rheum 1999;42:2471–78.PubMedCrossRef Elagib KEE, Tengnér P, Levi M, Jonsson R, Thompson K, Natvig JB, Wahren-Herlenius M. Immunoglobulin variable genes and epitope recognition of human monoclonal anti-Ro 52-kd in primary Sjögren’s syndrome. Arthritis Rheum 1999;42:2471–78.PubMedCrossRef
24.
go back to reference Pourmand N, Blange I, Ringertz N, Pettersson I. Intracellular localisation of the Ro 52kD auto-antigen in HeLa cells visualised with green fluorescent protein chimeras. Autoimmunity 1998;28:225–33.PubMedCrossRef Pourmand N, Blange I, Ringertz N, Pettersson I. Intracellular localisation of the Ro 52kD auto-antigen in HeLa cells visualised with green fluorescent protein chimeras. Autoimmunity 1998;28:225–33.PubMedCrossRef
25.
go back to reference Ottosson L, Hennig J, Espinosa A, Brauner S, Wahren-Herlenius M, Sunnerhagen M. Structural, functional and immunologic characterization of folded subdomains in the Ro52 protein targeted in Sjogren’s syndrome. Mol Immunol 2006;43:588–98.PubMedCrossRef Ottosson L, Hennig J, Espinosa A, Brauner S, Wahren-Herlenius M, Sunnerhagen M. Structural, functional and immunologic characterization of folded subdomains in the Ro52 protein targeted in Sjogren’s syndrome. Mol Immunol 2006;43:588–98.PubMedCrossRef
26.
go back to reference Lovgren T, Eloranta ML, Bave U, Alm GV, Ronnblom L. Induction of interferon-alpha production in plasmacytoid dendritic cells by immune complexes containing nucleic acid released by necrotic or late apoptotic cells and lupus IgG. Arthritis Rheum 2004;50:1861–72.PubMedCrossRef Lovgren T, Eloranta ML, Bave U, Alm GV, Ronnblom L. Induction of interferon-alpha production in plasmacytoid dendritic cells by immune complexes containing nucleic acid released by necrotic or late apoptotic cells and lupus IgG. Arthritis Rheum 2004;50:1861–72.PubMedCrossRef
27.
go back to reference Cederblad B, Blomberg S, Vallin H, Perers A, Alm GV, Ronnblom L. Patients with systemic lupus erythematosus have reduced numbers of circulating natural interferon-alpha- producing cells. J Autoimmun 1998;11:465–70.PubMedCrossRef Cederblad B, Blomberg S, Vallin H, Perers A, Alm GV, Ronnblom L. Patients with systemic lupus erythematosus have reduced numbers of circulating natural interferon-alpha- producing cells. J Autoimmun 1998;11:465–70.PubMedCrossRef
28.
go back to reference Pascual V, Farkas L, Banchereau J. Systemic lupus erythematosus: all roads lead to type I interferons. Curr Opin Immunol 2006;18:676–82.PubMedCrossRef Pascual V, Farkas L, Banchereau J. Systemic lupus erythematosus: all roads lead to type I interferons. Curr Opin Immunol 2006;18:676–82.PubMedCrossRef
29.
go back to reference Rönnblom L, Eloranta ML, Alm GV. The type I interferon system in systemic lupus erythematosus. Arthritis Rheum 2006;54:408–20.PubMedCrossRef Rönnblom L, Eloranta ML, Alm GV. The type I interferon system in systemic lupus erythematosus. Arthritis Rheum 2006;54:408–20.PubMedCrossRef
30.
go back to reference Rhodes DA, Ihrke G, Reinicke AT, Malcherek G, Towey M, Isenberg DA, Trowsdale J. The 52 000 MW Ro/SS-A autoantigen in Sjögren’s syndrome/systemic lupus erythematosus (Ro52) is an interferon-gamma inducible tripartite motif protein associated with membrane proximal structures. Immunology 2002;106:246–56.PubMedCrossRef Rhodes DA, Ihrke G, Reinicke AT, Malcherek G, Towey M, Isenberg DA, Trowsdale J. The 52 000 MW Ro/SS-A autoantigen in Sjögren’s syndrome/systemic lupus erythematosus (Ro52) is an interferon-gamma inducible tripartite motif protein associated with membrane proximal structures. Immunology 2002;106:246–56.PubMedCrossRef
31.
go back to reference Gerl V, Hostmann B, Johnen C, Waka A, Gerl M, Schumann F, Klein R, Radbruch A, Hiepe F. The intracellular 52-kd Ro/SSA autoantigen in keratinocytes is up-regulated by tumor necrosis factor alpha via tumor necrosis factor receptor I. Arthritis Rheum 2005;52:531–8.PubMedCrossRef Gerl V, Hostmann B, Johnen C, Waka A, Gerl M, Schumann F, Klein R, Radbruch A, Hiepe F. The intracellular 52-kd Ro/SSA autoantigen in keratinocytes is up-regulated by tumor necrosis factor alpha via tumor necrosis factor receptor I. Arthritis Rheum 2005;52:531–8.PubMedCrossRef
32.
go back to reference Rhodes DA, Trowsdale J. TRIM21 is a trimeric protein that binds IgG Fc via the B30.2 domain. Mol Immunol 2006;44:2406–14.PubMedCrossRef Rhodes DA, Trowsdale J. TRIM21 is a trimeric protein that binds IgG Fc via the B30.2 domain. Mol Immunol 2006;44:2406–14.PubMedCrossRef
33.
go back to reference Kong HJ, Anderson DE, Lee CH, Jang MK, Tamura T, Tailor P, Cho HK, Cheong J, Xiong H, Morse HC 3rd, Ozato K. Cutting edge: autoantigen Ro52 is an interferon inducible E3 ligase that ubiquitinates IRF-8 and enhances cytokine expression in macrophages. J Immunol 2007;179:26–30.PubMed Kong HJ, Anderson DE, Lee CH, Jang MK, Tamura T, Tailor P, Cho HK, Cheong J, Xiong H, Morse HC 3rd, Ozato K. Cutting edge: autoantigen Ro52 is an interferon inducible E3 ligase that ubiquitinates IRF-8 and enhances cytokine expression in macrophages. J Immunol 2007;179:26–30.PubMed
34.
go back to reference Sangfelt O, Erickson S, Grander D. Mechanisms of interferon-induced cell cycle arrest. Front Biosci 2000;5:D479–87.PubMedCrossRef Sangfelt O, Erickson S, Grander D. Mechanisms of interferon-induced cell cycle arrest. Front Biosci 2000;5:D479–87.PubMedCrossRef
35.
go back to reference Chawla-Sarkar M, Lindner DJ, Liu YF, Williams BR, Sen GC, Silverman RH, Borden EC. Apoptosis and interferons: role of interferon-stimulated genes as mediators of apoptosis. Apoptosis 2003;8:237–49.PubMedCrossRef Chawla-Sarkar M, Lindner DJ, Liu YF, Williams BR, Sen GC, Silverman RH, Borden EC. Apoptosis and interferons: role of interferon-stimulated genes as mediators of apoptosis. Apoptosis 2003;8:237–49.PubMedCrossRef
Metadata
Title
Interferon-α Induces Up-regulation and Nuclear Translocation of the Ro52 Autoantigen as Detected by a Panel of Novel Ro52-specific Monoclonal Antibodies
Authors
Linn Strandberg
Aurelie Ambrosi
Alexander Espinosa
Lars Ottosson
Maija-Leena Eloranta
Wei Zhou
Åse Elfving
Edward Greenfield
Vijay K. Kuchroo
Marie Wahren-Herlenius
Publication date
01-05-2008
Publisher
Springer US
Published in
Journal of Clinical Immunology / Issue 3/2008
Print ISSN: 0271-9142
Electronic ISSN: 1573-2592
DOI
https://doi.org/10.1007/s10875-007-9157-0

Other articles of this Issue 3/2008

Journal of Clinical Immunology 3/2008 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine