Skip to content
Licensed Unlicensed Requires Authentication Published by De Gruyter July 18, 2019

Only monospecific anti-DFS70 antibodies aid in the exclusion of antinuclear antibody associated rheumatic diseases: an Italian experience

  • Maria Infantino EMAIL logo , Francesca Pregnolato , Chelsea Bentow , Michael Mahler , Maurizio Benucci , Francesca Li Gobbi , Arianna Damiani , Valentina Grossi , Franco Franceschini , Caterina Bodio , Maria Orietta Borghi and Mariangela Manfredi

Abstract

Background

The dense fine speckled (DFS) is one of the most common patterns that can be observed as a result of the anti-nuclear antibodies (ANA) test on HEp-2 cells and is mostly caused by antibodies to DFS70 as the main antigenic target. As was recently demonstrated, isolated anti-DFS70 positivity can be used as an aid in the exclusion of ANA associated rheumatic diseases (AARD) due to the opportunity to better interpret unexplained positive IIF ANA results.

Methods

Our study included 333 subjects with AARD, 51 undifferentiated connective tissue disease (UCTD) patients, 235 disease controls and 149 healthy blood donors from an Italian cohort. All samples were tested for anti-DFS70 and anti-ENA antibodies using QUANTA Flash assays (Inova Diagnostics, San Diego, CA, USA).

Results

No differences in the prevalence of anti-DFS70 antibodies were seen among AARD, non-AARD and UCTD (2.1% [7/333] vs. 2.3% [9/384] vs. 5.9% [3/51], respectively; p-value = 0.188). AARD patients positive for anti-DFS70 antibodies showed in all cases an accompanying anti-ENA specificity. In contrast, monospecific anti-DFS70 antibodies showed a significantly different distribution with a clear trend across the main groups (AARD vs. non-AARD vs. UCTD: 0% [0/7] vs. 22% [2/9] vs. 100% [3/3], p = 0.007). Anti-DFS70 antibody levels among AARD, non-AARD and UCTD patients were not significantly different (p = 0.094). Within the anti-DFS70 antibody positive cases, AARD cohort showed a higher variability (median [min–max]: 3.2 [3.2–450.8] CU) compared to non-AARD (median [min–max]: 3.2 [3.2–75.7] CU) and UCTD patients (median [min–max]: 3.2 [3.2–59.0] CU).

Conclusions

Our preliminary data showed a similar frequency of anti-DFS70 antibodies in AARD, UCTD and non-AARD cohorts. Monospecificity of anti-DFS70 antibodies but not their mere presence is the key element in the diagnostic algorithm. Mono-specific anti-DFS70 antibodies might be a helpful biomarker to discriminate individuals with AARD from non-AARD presenting with a positive ANA.


Corresponding author: Dr. Maria Infantino, SOS Laboratorio Immunologia e Allegologia Ospedale S. Giovanni di Dio Firenze, Florence, Italy, Phone: +390556932502, Fax: +390556932289

aMaria Infantino and Francesca Pregnolato share 1st authorship.


  1. Author contributions: All the authors have accepted responsibility for the entire content of this submitted manuscript and approved submission.

  2. Research funding: None declared.

  3. Employment or leadership: None declared.

  4. Honorarium: None declared.

  5. Competing interests: The funding organization(s) played no role in the study design; in the collection, analysis, and interpretation of data; in the writing of the report; or in the decision to submit the report for publication.

References

1. Mahler M, Fritzler MJ. Epitope specificity and significance in systemic autoimmune diseases. Ann N Y Acad Sci 2010;1183:267–87.10.1111/j.1749-6632.2009.05127.xSearch in Google Scholar

2. Meroni PL, Schur PH. ANA screening: an old test with new recommendations. Ann Rheum Dis 2010;69:1420–2.10.1136/ard.2009.127100Search in Google Scholar

3. Fritzler MJ. The antinuclear antibody test: last or lasting gasp? Arthritis Rheum 2011;63:19–22.10.1002/art.30078Search in Google Scholar

4. Mahler M, Dervieux T. Comments on recent advances and recommendations for the assessment of autoantibodies to cellular antigens referred as antinuclear antibodies. Ann Rheum Dis 2014;73:e36.10.1136/annrheumdis-2014-205324Search in Google Scholar

5. Mahler M, Meroni PL, Bossuyt X, Fritzler MJ. Current concepts and future directions for the assessment of autoantibodies to cellular antigens referred to as anti-nuclear antibodies. J Immunol Res 2014;2014:315179.10.1155/2014/315179Search in Google Scholar

6. Ochs RL, Stein TW, Jr., Peebles CL, Gittes RF, Tan EM. Autoantibodies in interstitial cystitis. J Urol 1994;151:587–92.10.1016/S0022-5347(17)35023-1Search in Google Scholar

7. Conrad K, Rober N, Andrade LE, Mahler M. The clinical relevance of anti-DFS70 autoantibodies. Clin Rev Allergy Immunol 2017;52:202–16.10.1007/s12016-016-8564-5Search in Google Scholar

8. Ganapathy V, Daniels T, Casiano CA. LEDGF/p75: a novel nuclear autoantigen at the crossroads of cell survival and apoptosis. Autoimmun Rev 2003;2:290–7.10.1016/S1568-9972(03)00063-6Search in Google Scholar

9. Ochs RL, Muro Y, Si Y, Ge H, Chan EK, Tan EM. Autoantibodies to DFS 70 kd/transcription coactivator p75 in atopic dermatitis and other conditions. J Allergy Clin Immunol 2000;105:1211–20.10.1067/mai.2000.107039Search in Google Scholar PubMed

10. O’Rourke DJ, DiJohnson DA, Caiazzo RJ, Jr., Nelson JC, Ure D, O’Leary MP, et al. Autoantibody signatures as biomarkers to distinguish PCa from benign prostatic hyperplasia in patients with increased serum prostate specific antigen. Clin Chim Acta 2012;413:561–7.10.1016/j.cca.2011.11.027Search in Google Scholar PubMed PubMed Central

11. Mahler M, Parker T, Peebles CL, Andrade LE, Swart A, Carbone Y, et al. Anti-DFS70/LEDGF antibodies are more prevalent in healthy individuals compared to patients with systemic autoimmune rheumatic diseases. J Rheumatol 2012;39:2104–10.10.3899/jrheum.120598Search in Google Scholar

12. Albesa R, Sachs U, Infantino M, Manfredi M, Benucci M, Baus Y, et al. Increased prevalence of anti-DFS70 antibodies in young females: experience from a large international multi-center study on blood donors. Clin Chem Lab Med 2019;57:999–1005.10.1515/cclm-2018-1233Search in Google Scholar

13. Watanabe A, Kodera M, Sugiura K, Usuda T, Tan EM, Takasaki Y, et al. Anti-DFS70 antibodies in 597 healthy hospital workers. Arthritis Rheum 2004;50:892–900.10.1002/art.20096Search in Google Scholar

14. Shinohara T, Singh DP, Fatma N. LEDGF, a survival factor, activates stress-related genes. Prog Retin Eye Res 2000;21:341–58.10.1016/S1350-9462(02)00007-1Search in Google Scholar

15. Ge H, Si Y, Roeder RG. Isolation of cDNAs encoding novel transcription coactivators p52 and p75 reveals an alternate regulatory mechanism of transcriptional activation. EMBO J 1998;17:6723–9.10.1093/emboj/17.22.6723Search in Google Scholar PubMed PubMed Central

16. Daniels T, Zhang J, Gutierrez I, Elliot ML, Yamada B, Heeb MJ, et al. Antinuclear autoantibodies in prostate cancer: immunity to LEDGF/p75, a survival protein highly expressed in prostate tumors and cleaved during apoptosis. Prostate 2005;62:14–26.10.1002/pros.20112Search in Google Scholar PubMed

17. Maertens G, Cherepanov P, Pluymers W, Busschots K, De Clercq E, Debyser Z, et al. LEDGF/p75 is essential for nuclear and chromosomal targeting of HIV-1 integrase in human cells. J Biol Chem 2003;278:33528–39.10.1074/jbc.M303594200Search in Google Scholar PubMed

18. Muro Y, Ogawa Y, Sugiura K, Tomita Y. HLA-associated production of anti-DFS70/LEDGF autoantibodies and systemic autoimmune disease. J Autoimmun 2006;26:252–7.10.1016/j.jaut.2006.03.005Search in Google Scholar PubMed

19. Basu A, Drame A, Muñoz R, Gijsbers R, Debyser Z, De Leon M, et al. Pathway specific gene expression profiling reveals oxidative stress genes potentially regulated by transcription co-activator LEDGF/p75 in PCa cells. Prostate 2012;72:597–611.10.1002/pros.21463Search in Google Scholar PubMed PubMed Central

20. Singh DP, Ohguro N, Chylack LT, Jr., Shinohara T. Lens epithelium-derived growth factor: increased resistance to thermal and oxidative stresses. Invest Ophthalmol Vis Sci 1999;40:1444–51.Search in Google Scholar

21. Hendrix J, van Heertum B, Vanstreels E, Daelemans D, De Rijck J. Dynamics of the ternary complex formed by c-Myc interactor JPO2, transcriptional co-activator LEDGF/p75, and chromatin. J Biol Chem 2014;289:12494–506.10.1074/jbc.M113.525964Search in Google Scholar PubMed PubMed Central

22. Ochs RL, Mahler M, Basu A, Rios-Colon L, Sanchez TW, Andrade LE, et al. The significance of autoantibodies to DFS70/LEDGFp75 in health and disease: integrating basic science with clinical understanding. Clin Exp Med 2016;16:273–93.10.1007/s10238-015-0367-0Search in Google Scholar PubMed PubMed Central

23. Basu A, Sanchez TW, Casiano CA. DFS70/LEDGFp75: an enigmatic autoantigen at the interface between autoimmunity, AIDS, and cancer. Front Immunol 2015;6:116.10.3389/fimmu.2015.00116Search in Google Scholar PubMed PubMed Central

24. Debyser Z, Christ F, De Rijck J, Gijsbers R. Host factors for retroviral integration site selection. Trends Biochem Sci 2015;40: 108–16.10.1016/j.tibs.2014.12.001Search in Google Scholar PubMed

25. Basu A, Rojas H, Banerjee H, Cabrera IB, Perez KY, De Leon M, et al. Expression of the stress response oncoprotein LEDGF/p75 in human cancer: a study of 21 tumor types. PLoS One 2012;7:e30132.10.1371/journal.pone.0030132Search in Google Scholar PubMed PubMed Central

26. Bizzaro N, Tonutti E, Tampoia M, Infantino M, Cucchiaro F, Pesente F, et al. Specific chemoluminescence and immunoasdorption tests for anti-DFS70 antibodies avoid false positive results by indirect immunofluorescence. Clin Chim Acta 2015;451:271–7.10.1016/j.cca.2015.10.008Search in Google Scholar PubMed

27. Mahler M, Meroni PL, Andrade LE, Khamashta M, Bizzaro N, Casiano CA, et al. Towards a better understanding of the clinical association of anti-DFS70 autoantibodies. Autoimmun Rev 2016;15:198–201.10.1016/j.autrev.2015.11.006Search in Google Scholar PubMed

28. Bizzaro N, Pesente F, Cucchiaro F, Infantino M, Tampoia M, Villalta D, et al. Anti-DFS70 antibodies detected by immunoblot methods: a reliable tool to confirm the dense fine speckles ANA pattern. J Immunol Methods 2016;436:50–3.10.1016/j.jim.2016.06.008Search in Google Scholar PubMed

29. Muro Y, Sugiura K, Morita Y, Tomita Y. High concomitance of disease marker autoantibodies in anti-DFS70/LEDGF autoantibody-positive patients with autoimmune rheumatic disease. Lupus 2008;17:171–6.10.1177/0961203307086311Search in Google Scholar PubMed

30. Muro Y, Sugiura K, Nakashima R, Mimori T, Akiyama M. Low prevalence of anti-DFS70/LEDGF antibodies in patients with dermatomyositis and other systemic autoimmune rheumatic diseases. J Rheumatol 2013;40:92–3.10.3899/jrheum.121168Search in Google Scholar PubMed

31. Watanabe K, Muro Y, Sugiura K, Tomita Y. IgE and IgG(4) autoantibodies against DFS70/LEDGF in atopic dermatitis. Autoimmunity 2011;44:511–9.10.3109/08916934.2010.549157Search in Google Scholar PubMed

32. Miyara M, Albesa R, Charuel JL, El Amri M, Fritzler MJ, Ghillani-Dalbin P, et al. Clinical phenotypes of patients with anti-DFS70/LEDGF antibodies in a routine ANA referral cohort. Clin Dev Immunol 2013;2013:703759.10.1155/2013/703759Search in Google Scholar PubMed PubMed Central

33. Mahler M, Fritzler MJ. The clinical significance of the dense fine speckled immunofluorescence pattern on HEp-2 cells for the diagnosis of systemic autoimmune diseases. Clin Dev Immunol 2012;2012:494356.10.1155/2012/494356Search in Google Scholar PubMed PubMed Central

34. Rigon A, Buzzulini F, Soda P, Onofri L, Arcarese L, Iannello G, et al. Novel opportunities in automated classification of antinuclear antibodies on HEp-2 cell. Autoimmun Rev 2011;10:647–52.10.1016/j.autrev.2011.04.022Search in Google Scholar PubMed

35. Bentow C, Fritzler MJ, Mummert E, Mahler M. Recognition of the dense fine speckled (DFS) pattern remains challenging: results from an international internet-based survey. Auto Immun Highlights 2016;7:8.10.1007/s13317-016-0081-2Search in Google Scholar PubMed PubMed Central

36. Infantino M, Shovman O, Pérez D, Grossi V, Manfredi M, Benucci M, et al. A better definition of the anti-DFS70 antibody screening by IIF methods. Immunol Methods 2018;461:110–6.10.1016/j.jim.2018.07.001Search in Google Scholar PubMed

37. Malyavantham KS, Suresh L. Simultaneous distinction of monospecific and mixed DFS70 patterns during ANA screening with a novel HEp-2 ELITE/DFS70 knockout substrate. J Vis Exp 2018;17:131.10.3791/56722Search in Google Scholar PubMed PubMed Central

38. Malyavantham K, Suresh L. Analysis of DFS70 pattern and impact on ANA screening using a novel HEp-2 ELITE/DFS70 knockout substrate. Auto Immun Highlights 2017;8:3.10.1007/s13317-017-0091-8Search in Google Scholar PubMed PubMed Central

39. Bizzaro N, Fabris M. New genetically engineered DFS70 knock-out HEp-2 cells enable rapid and specific recognition of anti-DFS70 antibodies. Autoimmunity 2018;51:152–6.10.1080/08916934.2018.1469013Search in Google Scholar PubMed

40. Infantino M, Shovman O, Gilburd B, Manfredi M, Grossi V, Benucci M, et al. Improved accuracy in DFS pattern interpretation using a novel HEp-2 ELITE system. Clin Rheumatol 2019;38:1293–9.10.1007/s10067-018-04412-1Search in Google Scholar PubMed

41. Mahler M, Andrade LE, Casiano CA, Malyavantham K, Fritzler MJ. Anti-DFS70 antibodies: an update on our current understanding and their clinical usefulness. Expert Rev Clin Immunol 2019;15:241–50.10.1080/1744666X.2019.1562903Search in Google Scholar PubMed

42. Egerer K, Roggenbuck D, Hiemann R, Weyer MG, Büttner T, Radau B, et al. Automated evaluation of autoantibodies on human epithelial-2 cells as an approach to standardize cell-based immunofluorescence tests. Arthritis Res Ther 2010;12:R40.10.1186/ar2949Search in Google Scholar PubMed PubMed Central

43. Meroni PL, Bizzaro N, Cavazzana I, Borghi MO, Tincani A. Automated tests of ANA immunofluorescence as throughput autoantibody detection technology: strengths and limitations. BMC Med 2014;12:38.10.1186/1741-7015-12-38Search in Google Scholar PubMed PubMed Central

44. Fitch-Rogalsky C, Steber W, Mahler M, Lupton T, Martin L, Barr SG, et al. Clinical and serological features of patients referred through a rheumatology triage system because of positive antinuclear antibodies. PLoS One 2014;9:e93812.10.1371/journal.pone.0093812Search in Google Scholar PubMed PubMed Central

45. Mariz HA, Sato EI, Barbosa SH, Rodrigues SH, Dellavance A, Andrade LE. Pattern on the antinuclear antibody-HEp-2 test is a critical parameter for discriminating antinuclear antibody-positive healthy individuals and patients with autoimmune rheumatic diseases. Arthritis Rheum 2011;63:191–200.10.1002/art.30084Search in Google Scholar PubMed

46. Carter JB, Carter S, Saschenbrecker S, Goeckeritz BE. Recognition and relevance of anti-DFS70 autoantibodies in routine antinuclear autoantibodies testing at a community hospital. Front Med (Lausanne) 2018;5:88eb 11.10.3389/fmed.2018.00088Search in Google Scholar PubMed PubMed Central

47. Tedeschi SK, Johnson SR, Boumpas DT, Daikh D, Dörner T, Diamond B, et al. Multicriteria decision analysis process to develop new classification criteria for systemic lupus erythematosus. Ann Rheum Dis 2019 (in press).10.1136/annrheumdis-2018-214685Search in Google Scholar PubMed PubMed Central

48. Infantino M, Shovman O, Pérez D, Manfredi M, Grossi V, Benucci M, et al. Anti-DFS70 autoantibodies in undifferentiated connective tissue diseases subjects: what’s on the horizon? Rheumatology (Oxford) 2018 (in press).10.1093/rheumatology/key012Search in Google Scholar PubMed

49. Infantino M, Meacci F, Grossi V, Manfredi M, Li Gobbi F, Sarzi-Puttini PC, et al. The clinical impact of Anti-DFS70 antibodies in undifferentiated connective tissue disease: case reports and a review of the literature. Immunol Res 2017;65:293–5.10.1007/s12026-016-8836-4Search in Google Scholar PubMed

Received: 2019-04-30
Accepted: 2019-06-18
Published Online: 2019-07-18
Published in Print: 2019-10-25

©2019 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 23.4.2024 from https://www.degruyter.com/document/doi/10.1515/cclm-2019-0454/html
Scroll to top button