249
Views
18
CrossRef citations to date
0
Altmetric
ORIGINAL RESEARCH

Novel Th17 Lymphocyte Populations, Th17.1 and PD1+Th17, are Increased in Takayasu Arteritis, and Both Th17 and Th17.1 Sub-Populations Associate with Active Disease

, ORCID Icon, , ORCID Icon, , ORCID Icon, ORCID Icon, , , , , , , ORCID Icon & ORCID Icon show all
Pages 1521-1541 | Published online: 01 Mar 2022

References

  • Misra DP, Wakhlu A, Agarwal V, Danda D. Recent advances in the management of Takayasu arteritis. Int J Rheum Dis. 2019;22(Suppl 1):60–68. doi:10.1111/1756-185X.13285
  • Watts RA, Hatemi G, Burns JC, Mohammad AJ. Global epidemiology of vasculitis. Nat Rev Rheumatol. 2022;18(1):22–34. doi:10.1038/s41584-021-00718-8
  • Mirouse A, Biard L, Comarmond C, et al. Overall survival and mortality risk factors in Takayasu’s arteritis: a multicenter study of 318 patients. J Autoimmun. 2019;96:35–39. doi:10.1016/j.jaut.2018.08.001
  • Comarmond C, Biard L, Lambert M, et al. Long-term outcomes and prognostic factors of complications in Takayasu arteritis: a multicenter study of 318 patients. Circulation. 2017;136(12):1114–1122. doi:10.1161/CIRCULATIONAHA.116.027094
  • Misra DP, Rathore U, Patro P, Agarwal V, Sharma A. Patient-reported outcome measures in Takayasu arteritis: a systematic review and meta-analysis. Rheumatol Ther. 2021;8(3):1073–1093. doi:10.1007/s40744-021-00355-3
  • Arnaud L, Haroche J, Mathian A, Gorochov G, Amoura Z. Pathogenesis of Takayasu’s arteritis: a 2011 update. Autoimmun Rev. 2011;11(1):61–67. doi:10.1016/j.autrev.2011.08.001
  • Rathore U, Thakare DR, Patro P, Agarwal V, Sharma A, Misra DP. A systematic review of clinical and preclinical evidences for Janus kinase inhibitors in large vessel vasculitis. Clin Rheumatol. 2022;41(1):33–44. doi:10.1007/s10067-021-05973-4
  • Zhang H, Watanabe R, Berry GJ, Tian L, Goronzy JJ, Weyand CM. Inhibition of JAK-STAT signaling suppresses pathogenic immune responses in medium and large vessel vasculitis. Circulation. 2018;137(18):1934–1948. doi:10.1161/CIRCULATIONAHA.117.030423
  • Arend WP, Michel BA, Bloch DA, et al. The American College of Rheumatology 1990 criteria for the classification of Takayasu arteritis. Arthritis Rheum. 1990;33(8):1129–1134. doi:10.1002/art.1780330811
  • Jennette JC, Falk RJ, Bacon PA, et al. 2012 revised International Chapel Hill Consensus Conference Nomenclature of Vasculitides. Arthritis Rheum. 2013;65(1):1–11. doi:10.1002/art.37715
  • Misra DP, Misra R. Assessment of disease activity in Takayasu’s arteritis. Indian J Rheumatol. 2015;10(5):S43- S47. doi:10.1016/j.injr.2015.08.006
  • Uppal SS, Verma S. Analysis of the clinical profile, autoimmune phenomena and T cell subsets (CD4 and CD8) in Takayasu’s arteritis: a hospital-based study. Clin Exp Rheumatol. 2003;21(6 Suppl 32):S112–116.
  • Piggott K, Deng J, Warrington K, et al. Blocking the NOTCH pathway inhibits vascular inflammation in large-vessel vasculitis. Circulation. 2011;123(3):309–318. doi:10.1161/CIRCULATIONAHA.110.936203
  • Tian Y, Li J, Tian X, Zeng X. Using the co-expression network of T cell-activation-related genes to assess the disease activity in Takayasu’s arteritis patients. Arthritis Res Ther. 2021;23(1):303. doi:10.1186/s13075-021-02636-2
  • Saadoun D, Garrido M, Comarmond C, et al. Th1 and Th17 cytokines drive inflammation in Takayasu arteritis. Arthritis Rheumatol. 2015;67(5):1353–1360. doi:10.1002/art.39037
  • Misra DP, Chaurasia S, Misra R. Increased circulating Th17 cells, serum IL-17A, and IL-23 in Takayasu Arteritis. Autoimmune Dis. 2016;2016:7841718. doi:10.1155/2016/7841718
  • Tesmer LA, Lundy SK, Sarkar S, Fox DA. Th17 cells in human disease. Immunol Rev. 2008;223:87–113. doi:10.1111/j.1600-065X.2008.00628.x
  • Zúñiga LA, Jain R, Haines C, Cua DJ. Th17 cell development: from the cradle to the grave. Immunol Rev. 2013;252(1):78–88. doi:10.1111/imr.12036
  • Ramesh R, Kozhaya L, McKevitt K, et al. Pro-inflammatory human Th17 cells selectively express P-glycoprotein and are refractory to glucocorticoids. J Exp Med. 2014;211(1):89–104. doi:10.1084/jem.20130301
  • Bordon Y. T cells: spotting the troublemakers. Nat Rev Immunol. 2014;14(2):64–65. doi:10.1038/nri3610
  • Prasad S, Tripathi D, Rai MK, Aggarwal S, Mittal B, Agarwal V. Multidrug resistance protein-1 expression, function and polymorphisms in patients with rheumatoid arthritis not responding to methotrexate. Int J Rheum Dis. 2014;17(8):878–886. doi:10.1111/1756-185X.12362
  • Kansal A, Tripathi D, Rai MK, Agarwal V. Persistent expression and function of P-glycoprotein on peripheral blood lymphocytes identifies corticosteroid resistance in patients with systemic lupus erythematosus. Clin Rheumatol. 2016;35(2):341–349. doi:10.1007/s10067-015-3079-7
  • Edavalath S, Rai MK, Gupta V, et al. Tacrolimus induces remission in refractory and relapsing lupus nephritis by decreasing P‐glycoprotein expression and function on peripheral blood lymphocytes. Rheumatol Int. 2022. Epub ahead of print. doi:10.1007/s00296-021-05057-1
  • Tristão FSM, Rocha FA, Carlos D, et al. Th17-inducing cytokines IL-6 and IL-23 are crucial for granuloma formation during experimental paracoccidioidomycosis. Front Immunol. 2017;8:949. doi:10.3389/fimmu.2017.00949
  • Ramstein J, Broos CE, Simpson LJ, et al. IFN-γ-producing T-Helper 17.1 cells are increased in sarcoidosis and are more prevalent than T-Helper Type 1 cells. Am J Respir Crit Care Med. 2016;193(11):1281–1291. doi:10.1164/rccm.201507-1499OC
  • Broos CE, Koth LL, van Nimwegen M, et al. Increased T-helper 17.1 cells in sarcoidosis mediastinal lymph nodes. Eur Respir J. 2018;51:3. doi:10.1183/13993003.01124-2017
  • Jiang Y, Li Y, Zhu B. T-cell exhaustion in the tumor microenvironment. Cell Death Dis. 2015;6(6):. doi:10.1038/cddis.2015.162.
  • Celada LJ, Kropski JA, Herazo-Maya JD, et al. PD-1 up-regulation on CD4(+) T cells promotes pulmonary fibrosis through STAT3-mediated IL-17A and TGF-β1 production. Sci Transl Med. 2018;10:460. doi:10.1126/scitranslmed.aar8356
  • Stojanovic M, Raskovic S, Milivojevic V, et al. Enhanced liver fibrosis score as a biomarker for vascular damage assessment in patients with takayasu arteritis-a pilot study. J Cardiovasc Dev Dis. 2021;8(12):187. doi:10.3390/jcdd8120187
  • Li T, Gao N, Cui W, et al. The role of CD8(+) Granzyme B(+) T cells in the pathogenesis of Takayasu’s arteritis. Clin Rheumatol. 2021;41(1):167. doi:10.1007/s10067-021-05903-4.
  • Gao N, Cui W, Zhao LM, Li TT, Zhang JH, Pan LL. Contribution of Th2-like Treg cells to the pathogenesis of Takayasu’s arteritis. Clin Exp Rheumatol. 2020;38 Suppl 124(2):48–54.
  • Abdallah HM, Al-Abd AM, El-Dine RS, El-Halawany AM. P-glycoprotein inhibitors of natural origin as potential tumor chemo-sensitizers: a review. J Adv Res. 2015;6(1):45–62. doi:10.1016/j.jare.2014.11.008
  • Lum BL, Gosland MP. MDR expression in normal tissues: pharmacologic implications for the clinical use of P-glycoprotein inhibitors. Hematol Oncol Clin North Am. 1995;9(2):319–336. doi:10.1016/S0889-8588(18)30097-2
  • Jain A, Singh H, Nath A, et al. Distinct T-cell immunophenotypic signature in a subset of sarcoidosis patients with arthritis. J R Coll Physicians Edinb. 2020;50(3):226–232. doi:10.4997/JRCPE.2020.304
  • Misra R, Danda D, Rajappa SM, et al. Development and initial validation of the Indian Takayasu Clinical Activity Score (ITAS2010). Rheumatology (Oxford). 2013;52(10):1795–1801. doi:10.1093/rheumatology/ket128
  • Campochiaro C, Misra DP. PET in Takayasu arteritis: onwards and upwards towards a future of robust multimodality disease activity assessment? Rheumatology (Oxford). 2021. doi:10.1093/rheumatology/keab644
  • Singh H, Agarwal V, Chaturvedi S, Misra DP, Jaiswal AK, Prasad N. Reciprocal relationship between HDAC2 and P-Glycoprotein/MRP-1 and their role in steroid resistance in childhood nephrotic syndrome. Front Pharmacol. 2019;10:558. doi:10.3389/fphar.2019.00558
  • Sample size calculator using means. Available from: https://sample-size.net/sample-size-means/. Accessed 19 December 2021.
  • Hata A, Noda M, Moriwaki R, Numano F. Angiographic findings of Takayasu arteritis: new classification. Int J Cardiol. 1996;54(Suppl):S155–163. doi:10.1016/S0167-5273(96)02813-6
  • Goel R, Kabeerdoss J, Ram B, et al. Serum cytokine profile in asian Indian patients with takayasu arteritis and its association with disease activity. Open Rheumatol J. 2017;11:23–29. doi:10.2174/1874312901711010023
  • Jenkins DG, Quintana-Ascencio PF. A solution to minimum sample size for regressions. PLoS One. 2020;15(2):e0229345. doi:10.1371/journal.pone.0229345
  • Miroux C, Morales O, Ghazal K, et al. In vitro effects of cyclosporine A and tacrolimus on regulatory T-cell proliferation and function. Transplantation. 2012;94(2):123–131. doi:10.1097/TP.0b013e3182590d8f
  • Deng J, Younge BR, Olshen RA, Goronzy JJ, Weyand CM. Th17 and Th1 T-cell responses in giant cell arteritis. Circulation. 2010;121(7):906–915. doi:10.1161/CIRCULATIONAHA.109.872903
  • Suzuki K, Saito K, Tsujimura S, et al. Tacrolimus, a calcineurin inhibitor, overcomes treatment unresponsiveness mediated by P-glycoprotein on lymphocytes in refractory rheumatoid arthritis. J Rheumatol. 2010;37(3):512–520. doi:10.3899/jrheum.090048
  • Chaturvedi S, Rai M, Singh H, et al. Dual inhibition by phosphodiesterase 5 and 5-HT2B Inhibitor leads to near complete amelioration of fibrotic potential of human adult dermal fibroblasts isolated from a scleroderma patient. Indian J Rheumatol. 2021;16(1):43–48. doi:10.4103/injr.injr_169_19
  • Misra DP, Sharma A, Kadhiravan T, Negi VS. A scoping review of the use of non-biologic disease modifying anti-rheumatic drugs in the management of large vessel vasculitis. Autoimmun Rev. 2017;16(2):179–191. doi:10.1016/j.autrev.2016.12.009
  • Langford CA, Cuthbertson D, Ytterberg SR, et al. A randomized, double-blind trial of Abatacept (CTLA-4Ig) for the treatment of Takayasu Arteritis. Arthritis Rheumatol. 2017;69(4):846–853. doi:10.1002/art.40037
  • Nakaoka Y, Isobe M, Takei S, et al. Efficacy and safety of tocilizumab in patients with refractory Takayasu arteritis: results from a randomised, double-blind, placebo-controlled, Phase 3 trial in Japan (the TAKT study). Ann Rheum Dis. 2018;77(3):348–354. doi:10.1136/annrheumdis-2017-211878
  • Misra DP, Rathore U, Patro P, Agarwal V, Sharma A. Disease-modifying anti-rheumatic drugs for the management of Takayasu arteritis—a systematic review and meta-analysis. Clin Rheumatol. 2021;40(11):4391–4416. doi:10.1007/s10067-021-05743-2
  • Singh H, Prasad N, Misra DP, Jaiswal AK, Agarwal V. P-glycoprotein and/or histone deacetylase 2 regulates steroid responsiveness in childhood nephrotic syndrome. Indian J Rheumatol. 2020;15(1):5–10. doi:10.4103/injr.injr_126_19
  • Acharya N, Sharma SK, Mishra D, Dhooria S, Dhir V, Jain S. Efficacy and safety of pirfenidone in systemic sclerosis-related interstitial lung disease-a randomised controlled trial. Rheumatol Int. 2020;40(5):703–710. doi:10.1007/s00296-020-04565-w
  • Distler O, Highland KB, Gahlemann M, et al. Nintedanib for systemic sclerosis–associated interstitial lung disease. N Engl J Med. 2019;380(26):2518–2528. doi:10.1056/NEJMoa1903076
  • Khanna D, Lin CJF, Furst DE, et al. Tocilizumab in systemic sclerosis: a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Respir Med. 2020;8(10):963–974. doi:10.1016/S2213-2600(20)30318-0
  • Lammas DA, De Heer E, Edgar JD, et al. Heterogeneity in the granulomatous response to mycobacterial infection in patients with defined genetic mutations in the interleukin 12-dependent interferon-gamma production pathway. Int J Exp Pathol. 2002;83(1):1–20. doi:10.1046/j.1365-2613.2002.00216.x
  • Arkema EV, Cozier YC. Epidemiology of sarcoidosis: current findings and future directions. Ther Adv Chronic Dis. 2018;9(11):227–240. doi:10.1177/2040622318790197
  • Gonzalez-Gay MA, Martinez-Dubois C, Agudo M, Pompei O, Blanco R, Llorca J. Giant cell arteritis: epidemiology, diagnosis, and management. Curr Rheumatol Rep. 2010;12(6):436–442. doi:10.1007/s11926-010-0135-9
  • Hoffman GS. Takayasu arteritis: lessons from the American National Institutes of Health experience. Int J Cardiol. 1996;54(Suppl):S99–102. doi:10.1016/S0167-5273(96)88778-X
  • Incerti E, Tombetti E, Fallanca F, et al. 18F-FDG PET reveals unique features of large vessel inflammation in patients with Takayasu’s arteritis. Eur J Nucl Med Mol Imaging. 2017;44(7):1109–1118. doi:10.1007/s00259-017-3639-y
  • Adriawan IR, Atschekzei F, Dittrich-Breiholz O, et al. Novel aspects of regulatory T cell dysfunction as a therapeutic target in giant cell arteritis. Ann Rheum Dis. 2022;81(1):124–131. doi:10.1136/annrheumdis-2021-220955