Ankylosing spondylitis: genetic aspects

DOI: https://doi.org/10.29296/25877305-2019-12-19
Download full text PDF
Issue: 
12
Year: 
2019

V. Mordovsky(1, 2), Candidate of Medical Sciences; E. Kapustina(1, 2), Candidate of Medical Sciences; A. Chernova(1, 2), MD; S. Nikulina(1), MD; N. Aksyutina(1), MD; T. Potupchik(1),Candidate of Medical Sciences 1-Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University 2-Russian-Italian Laboratory of Medical Genetics MAGI Russia, Krasnoyarsk

The paper presents a review of the literature on ankylosing spondylitis (AS), a chronic inflammatory autoimmune disease predominantly involving the axial skeleton. Since the beginning of the era of the genome-wide association studies (GWAS), more than 150 loci associated with AS have been identified, but this can explain no more than 30% of inheritance cases. The review attempts to identify the role of the presented genes in the development of AS. To continue investigations in the above direction will be able to create prerequisites for the emergence of new goals for targeted therapy.

Keywords: 
genetics
ankylosing spondylitis
HLA-B27
TNF
ERAP1
single nucleotide polymorphism
genetics



It appears your Web browser is not configured to display PDF files. Download adobe Acrobat или click here to download the PDF file.

References: 
  1. Li Z., Haynes K., Pennisi D. et al. Epigenetic and gene expression analysis of ankylosing spondylitis-associated loci implicate immune cells and the gut in the disease pathogenesis // Genes and Immunity. – 2017; 18 (3): 135–43. DOI: 10.1038/gene.2017.11.
  2. Ranganathan V., Gracey E., Brown M. et al. Pathogenesis of ankylosing spondylitis – recent advances and future directions // Nat. Rev. Rheumatol. – 2017; 13 (6): 359–67. DOI: 10.1038/nrrheum.2017.56.
  3. Brown M., Kenna T., Wordsworth B. Genetics of ankylosing spondylitis - insights into pathogenesis // Nat. Rev. Rheumatol. – 2015; 12 (2): 81–91. DOI: 10.1038/nrrheum.2015.133.
  4. Ranganathan V., Gracey E., Brown M. et al. Pathogenesis of ankylosing spondylitis - recent advances and future directions // Nat. Rev. Rheumatol. – 2017; 13 (6): 359–67. DOI: 10.1038/nrrheum.2017.56.
  5. Hanson A., Brown M. Genetics and the Causes of Ankylosing Spondylitis. // Rheum. Dis. Clin. North Am. – 2017; 43 (3): 401–14. DOI: 10.1016/j.rdc.2017.04.006.
  6. Dashti N., Mahmoudi M., Aslani S. et al. HLA-B*27 subtypes and their implications in the pathogenesis of ankylosing spondylitis // Gene. – 2018; 670: 15–21. DOI: 10.1016/j.gene.2018.05.092.
  7. Colbert R., Tran T., Layh-Schmitt G. HLA-B27 misfolding and ankylosing spondylitis // Mol. Immunol. – 2014; 57 (1): 44–51. DOI: 10.1016/j.molimm.2013.07.013.
  8. Dashti N., Mahmoudi M., Aslani S. et al. HLA-B*27 subtypes and their implications in the pathogenesis of ankylosing spondylitis // Gene. – 2018; 670: 15–21. DOI: 10.1016/j.gene.2018.05.092.
  9. Schittenhelm R., Sian T., Wilmann P. et al. Revisiting the Arthritogenic Peptide Theory: Quantitative Not Qualitative Changes in the Peptide Repertoire of HLA-B27 Allotypes // Arthritis & Rheumatology. – 2015; 67 (3): 702–13. DOI: 10.1002/art.38963.
  10. Lin H., Gong Y.-Z. Association of HLA-B27 with ankylosing spondylitis and clinical features of the HLA-B27-associated ankylosing spondylitis: a meta-analysis // Rheumatol. Int. – 2017; 37 (8): 1267–80. DOI: 10.1007/s00296-017-3741-2.
  11. Cortes A., Pulit S., Leo P. et al. Major histocompatibility complex associations of ankylosing spondylitis are complex and involve further epistasis with ERAP1 // Nature Communications. – 2015; 6 (1): 3–11. DOI: 10.1038/ncomms8146.
  12. Li H., Li Q., Ji C. et al. Ankylosing Spondylitis Patients with HLA-B*2704 have More Uveitis than Patients with HLA-B*2705 in a North Chinese Population // Ocular Immunol. Inflamm. – 2016; 26 (1): 65–9. DOI: 10.1080/09273948.2016.1188967.
  13. Chen L., Shi H., Yuan J. et al. Position 97 of HLA-B, a residue implicated in pathogenesis of ankylosing spondylitis, plays a key role in cell surface free heavy chain expression // Ann. Rheum. Dis. – 2016; 76 (3): 593–601. DOI: 10.1136/annrheumdis-2016-209512.
  14. Li Z., Brown M. Progress of genome-wide association studies of ankylosing spondylitis. // Clin. Transl. Immunol. – 2017; 6 (12): e163. DOI: 10.1038/cti.2017.49.
  15. Kenna T., Robinson P., Haroon N. Endoplasmic reticulum aminopeptidases in the pathogenesis of ankylosing spondylitis // Rheumatology. – 2015; 54 (9): 1549–56. DOI: 10.1093/rheumatology/kev218.
  16. Meng Q., Zhang X., Liu X. et al. Association of PTPN22 polymorphsims and ankylosing spondylitis susceptibility // Int. J. Clin. Experim. Pathol. – 2015; 8 (1): 933–7.
  17. Lee Y., Song G. Associations between ERAP1 polymorphisms and susceptibility to ankylosing spondylitis: a meta-analysis // Clin. Rheumatol. – 2016; 35 (8): 2009–15. DOI: 10.1007/s10067-016-3287-9.
  18. Martin-Esteban A., Sanz-Bravo A., Guasp P. et al. Separate effects of the ankylosing spondylitis associated ERAP1 and ERAP2 aminopeptidases determine the influence of their combined phenotype on the HLA-B*27 peptidome // J. Autoimmun. – 2017; 79 (4): 28–38. DOI: 10.1016/j.jaut.2016.12.008.
  19. Andrés A., Dennis M., Kretzschmar W. et al. Balancing selection maintains a form of ERAP2 that undergoes nonsense-mediated decay and affects antigen presentation // PLoS Genet. – 2010; 6 (10): e1001157. DOI: 10.1371/journal.pgen.1001157.
  20. Robinson P., Costello M., Leo P. et al. ERAP2 is associated with ankylosing spondylitis inHLA-B27-positive and HLA-B27-negative patients // Ann. Rheum. Dis. – 2015; 74 (8): 1627–9. DOI: 10.1136/annrheumdis-2015-207416.
  21. Raychaudhuri S., Raychaudhuri S. IL-23/IL-17 axis in spondyloarthritis-bench to bedside // Clin. Rheumatol. – 2016; 35 (6): 1437–41. DOI: 10.1007/s10067-016-3263-4.
  22. Cortes A., Hadler J., Pointon J. et al. Identification of multiple risk variants for ankylosing spondylitis through high-density genotyping of immune-related loci // Nat. Gen. – 2013; 45: 730–8.
  23. Ghoreschi K., Laurence A., Yang X. et al. Generation of pathogenic T(H)17 cells in the absence of TGF-beta signaling // Nature. – 2010; 467: 967–71. DOI: 10.1038/nature09447.
  24. Lee Y., Song G. Associations between interleukin-23R polymorphisms and ankylosing spondylitis susceptibility: an updated meta-analysis // Zeitschrift für Rheumatologie. – 2018; 78 (3): 272–80. DOI: 10.1007/s00393-018-0472-z.
  25. Vidal-Castiñeira J., López-Vázquez A., Diaz-Peña R. et al. A Single Nucleotide Polymorphism in the Il17ra Promoter Is Associated with Functional Severity of Ankylosing Spondylitis // Public Library of Science. – 2016; 11 (7): e0158905. DOI: 10.1371/journal.pone.0158905.
  26. Zhang L., Fan D., Liu L. et al. Association Study of IL-12B Polymorphisms Susceptibility with Ankylosing Spondylitis in Mainland Han Population // Public Library of Science. – 2015; 10 (6): e0130982. DOI: 10.1371/journal.pone.0130982.
  27. Yang B., Xu Y., Liu X. et al. IL-23R and IL-17A polymorphisms correlate with susceptibility of ankylosing spondylitis in a Southwest Chinese population // Oncotarget. – 2017; 8 (41): 70310–6. DOI: 10.18632/oncotarget.20319.
  28. Ruan W., Xie J., Jin Q. et al. The Diagnostic and Prognostic Role of Interleukin 12B and Interleukin 6R Gene Polymorphism in Patients With Ankylosing Spondylitis // J. Clin. Rheumatol. – 2017; 24 (1): 18–24. DOI: 10.1097/rhu.0000000000000610.
  29. Kaplanski G. Interleukin-18: Biological properties and role in disease pathogenesis // Immunol. Rev. – 2017; 281 (1): 138–53. DOI: 10.1111/imr.12616.
  30. Manolova I., Ivanova M., Boyadzhieva V. et al. Elevated serum levels of IL-18 in patients with rheumatoid arthritis and ankylosing spondylitis // Revmatologiia (Bulgaria). – 2013; 21 (2): 35–41.
  31. Ivanova M., Manolova I., Goycheva P. et al. Serum cytokines (TNF-alpha and IL-18) in ankylosing spondylitis in relation to disease activity // Comptes rendus de l’Académie bulgare des sciences: sciences mathématiques et naturelles. – 2014; 67 (4): 593–602.
  32. Sode J., Bank S., Vogel U. et al. Genetically determined high activities of the TNF-alpha, IL23/IL17, and NFkB pathways were associated with increased risk of ankylosing spondylitis. // BMC Med. Genet. – 2018; 19 (1): 165. DOI: 10.1186/s12881-018-0680-z.
  33. Wang W., Liu Y., Ma X. et al. Association Between Protein Tyrosine Phosphatase Non-Receptor Type 22 (PTPN22) Polymorphisms and Risk of Ankylosing Spondylitis: A Meta-analysis // Med. Sci. Monit. – 2017; 23: 2619–24. DOI: 10.12659/MSM.901083.
  34. Meng Q., Zhang X., Liu X. et al. Association of PTPN22 polymorphsims and ankylosing spondylitis susceptibility // Int. J. Clin. Experim. Pathol. – 2015; 8 (1): 933–7.
  35. El-Lebedy D., Raslan H., Ibrahim A. et al. Association of STAT4 rs7574865 and PTPN22 rs2476601 polymorphisms with rheumatoid arthritis and non-systemically reacting antibodies in Egyptian patients // Clin Rheumatol. – 2017; 36 (9): 1981–7. DOI: 10.1007/s10067-017-3632-7.
  36. Dahmani C., Benzaoui A., Amroun H. et al. Association of the HLA-B27 antigen and the CTLA4 gene CT60/rs3087243 polymorphism with ankylosing spondylitis in Algerian population: A case-control study // Int. J. Immunogen. – 2018; 45 (3): 109–17. DOI: 10.1111/iji.12369.
  37. Wu J., Zhang L., Zhou Y. The association between CTLA-4 (+49 A/G) polymorphism and susceptibility to ankylosing spondylitis: a meta-analysis // Int. J. Rheum. Dis. – 2015; 19 (12): 1237–43. http://dx.doi.org/10.1111/1756-185x.12705.
  38. Huang C.-H., Wei J., Chen C.-C. et al. Associations of the PTPN22 and CTLA-4 genetic polymorphisms with Taiwanese ankylosing spondylitis // Rheumatol. Int. – 2014; 34 (5): 683–91. DOI: 10.1007/s00296-013-2894-x.
  39. Guseva I.A., Luchihina E.L., Abramov D.D. i dr. Polimorfizm gena CTLA-4 (+49A>G) – prediktor rannego naznachenija genno-inzhenernyh biologicheskih preparatov u bol'nyh rannim revmatoidnym artritom, ne otvechajuschih na terapiju metotreksatom. Molekuljarnaja diagnostika, 2017. Sb. tr. IX Vseross. nauchno-prakt. konf. s mezhdunar. uchastiem. 2017, 526–7 [Guseva I.A., Luchikhina E.L., Abramov D.D. et al. Polimorfizm gena CTLA-4 (+49A>G) – prediktor rannego naznacheniya genno- inzhenernykh biologicheskikh preparatov u bol’nykh rannim revmatoidnym artritom, ne otvechayushchikh na terapiyu metotreksatom. Molekulyarnaya diagnostika, 2017. Sb. Tr. IX Vseross. nauchno-prakt. konf. s mezhdunar. uchastiem. 2017; 526–7 (in Russ.)].
  40. Ntusi N., Francis J., Sever E. et al. Anti-TNF modulation reduces myocardial inflammation and improves cardiovascular function in systemic rheumatic diseases // Int. J. Cardiol. – 2018; 270: 253–9. DOI: 10.1016/j.ijcard.2018.06.099.
  41. Hu N., Cui Y., Yang Q. et al. Association of polymorphisms in TNF and GRN genes with ankylosing spondylitis in a Chinese Han population // Rheumatol. Int. – 2018; 38 (3): 481–7. DOI: 10.1007/s00296-017-3899-7.