Turn off MathJax
Article Contents
Xinju Wang, Jianru Chen, Wei Wu, Jinrong Fan, Luling Huang, Weiwei Sun, Kaiqiao He, Shuli Li, Chunying Li. Circulating Memory T Cells and TCF1+ T Cells Aid in Diagnosis and Monitor Disease Activity in Vitiligo[J]. Journal of Pharmaceutical Analysis. doi: 10.1016/j.jpha.2024.100998
Citation: Xinju Wang, Jianru Chen, Wei Wu, Jinrong Fan, Luling Huang, Weiwei Sun, Kaiqiao He, Shuli Li, Chunying Li. Circulating Memory T Cells and TCF1+ T Cells Aid in Diagnosis and Monitor Disease Activity in Vitiligo[J]. Journal of Pharmaceutical Analysis. doi: 10.1016/j.jpha.2024.100998

Circulating Memory T Cells and TCF1+ T Cells Aid in Diagnosis and Monitor Disease Activity in Vitiligo

doi: 10.1016/j.jpha.2024.100998
Funds:

This study was funded by National Natural Science Foundation of China (No. 82222059, No. 82173416, and No. 81930087).

  • Received Date: Dec. 19, 2023
  • Accepted Date: May 05, 2024
  • Rev Recd Date: Mar. 28, 2024
  • Available Online: May 10, 2024
  • Vitiligo is an immune memory skin disease. T-cell factor 1 (TCF1) is essential for maintaining the maintenance of the memory T-cell pool. There is an urgent need to investigate the characteristics of peripheral memory T-cell profile and TCF1+ T-cell frequencies in patients with vitiligo patients. In this study, 31 patients with active vitiligo (AV), 22 with stable vitiligo (SV), and 30 healthy controls (HCs) were included. We measured circulating memory and TCF1+ T-cell frequencies using by flow cytometry. The Spearman’s rank test was used to evaluate the correlation correlations between cell frequencies and disease characteristics. Receiver operating characteristic curves (ROC) were constructed to investigate the discriminative power of the cell subpopulations. Circulating CD4+ and CD8+ terminally differentiated effector memory T-cell (TEMRA) frequencies were significantly higher in the AV group than in compared to HCs (P < 0.05). TCF1+ T-cell subpopulations were widespread increased in patients with vitiligo patients (P < 0.05). After adjusting adjustment for potential confounders, CD8+ and CD4+ central memory (TCM) cells, and CD8+ TEMRA were correlated with disease activity (P < 0.05). The combined diagnostic value of the four (naïve naive, effector memory, TCM, and TEMRA) CD8+TCF1+ T-cell subsets was relatively high (area under the ROC curve [AUC]AUC = 0.804, sensitivity = 71.70%, specificity = 83.34%), and the CD8+ T-cell subsets combination performed well in discriminating disease activity (AUC = 0.849, sensitivity = 70.97%, specificity = 90.91%). We demonstrated an altered circulating memory T-cell profile and increased TCF1+ T-cell percentage percentages in patients with vitiligo patients. T-cell subpopulations had a strong value values for vitiligo diagnosis and activity evaluation. This evidence presents a potential new pharmacological target for inhibiting to inhibit the autoimmunity that leads leading to vitiligo disease activity.
  • loading
  • [1]
    K. Ezzedine, V. Eleftheriadou, M. Whitton, et al., Vitiligo, Lancet 386 (2015) 74-84.
    [2]
    M.W. Linthorst Homan, P.I. Spuls, J. de Korte, et al., The burden of vitiligo: Patient characteristics associated with quality of life, J. Am. Acad. Dermatol. 61 (2009) 411-420.
    [3]
    K. Boniface, J. Seneschal, M. Picardo, et al., Vitiligo: Focus on clinical aspects, immunopathogenesis, and therapy, Clin. Rev. Allergy. Immunol. 54 (2018) 52-67.
    [4]
    M. Cavalie, K. Ezzedine, E. Fontas, et al., Maintenance therapy of adult vitiligo with 0.1% tacrolimus ointment: A randomized, double blind, placebo-controlled study, J. Invest. Dermatol. 135 (2015) 970-974.
    [5]
    J.M. Richmond, J.P. Strassner, M. Rashighi, et al., Resident memory and recirculating memory T cells cooperate to maintain disease in a mouse model of vitiligo, J. Invest. Dermatol. 139 (2019) 769-778.
    [6]
    R.L. Riding, J.E. Harris, The Role of Memory CD8+ T Cells in Vitiligo, J. Immunol. 203 (2019) 11-19.
    [7]
    K. Boniface, C. Jacquemin, A.S. Darrigade, et al., Vitiligo skin is imprinted with resident memory CD8 T cells expressing CXCR3, J. Invest. Dermatol. 138 (2018) 355-364.
    [8]
    C. Martins, A.S. Darrigade, C. Jacquemin, et al., Phenotype and function of circulating memory T cells in human vitiligo, Br. J. Dermatol. 183 (2020) 899-908.
    [9]
    X. Zhao, Q. Shan, H.H. Xue, TCF1 in T cell immunity: A broadened frontier, Nat. Rev. Immunol. 22 (2022) 147-157.
    [10]
    I. Siddiqui, K. Schaeuble, V. Chennupati, et al., Intratumoral Tcf1+PD-1+CD8+ T cells with stem-like properties promote tumor control in response to vaccination and checkpoint blockade immunotherapy, Immunity. 50 (2019) 195-211.e10.
    [11]
    B.C. Miller, D.R. Sen, R. Al Abosy, et al., Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade, Nat. Immunol. 20 (2019) 326-336.
    [12]
    R.L. Rutishauser, C.D.T. Deguit, J. Hiatt, et al., TCF-1 regulates HIV-specific CD8+ T cell expansion capacity, JCI Insight 6 (2021), e136648.
    [13]
    R. Maniar, P.H. Wang, R.S. Washburn, et al., Self-renewing CD8+ T-cell abundance in blood associates with response to immunotherapy, Cancer Immunol. Res. 11 (2023) 164-170.
    [14]
    L.M. Snell, B.L. MacLeod, J.C. Law, et al., CD8+ T cell priming in established chronic viral infection preferentially directs differentiation of memory-like cells for sustained immunity, Immunity 49 (2018) 678-694.e5.
    [15]
    S.V. Gearty, F. Dundar, P. Zumbo, et al., An autoimmune stem-like CD8 T cell population drives type 1 diabetes, Nature 602 (2022) 156-161.
    [16]
    T. Lei, A. Xu, T. Gao, et al., Consensus on the diagnosis and treatment of vitiligo in china (2021 Revision), Int. J. Dermatol. Venereol. 4 (2021) 10-15.
    [17]
    G.R. Kanthraj, C.R. Srinivas, S.D. Shenoi, et al., Comparison of computer-aided design and rule of nines methods in the evaluation of the extent of body involvement in cutaneous lesions, Arch. Dermatol. 133 (1997) 922-923.
    [18]
    F. Sallusto, D. Lenig, R. Forster, et al., Two subsets of memory T lymphocytes with distinct homing potentials and effector functions, Nature 401 (1999) 708-712.
    [19]
    J.M. Richmond, J.P. Strassner, K.I. Essien, et al., T-cell positioning by chemokines in autoimmune skin diseases, Immunol. Rev. 289 (2019) 186-204.
    [20]
    F. Shah, S. Patel, R. Begum, et al., Emerging role of tissue resident memory T cells in vitiligo: From pathogenesis to therapeutics, Autoimmun. Rev. 20 (2021), 102868.
    [21]
    F. Shah, P.S. Giri, A.H. Bharti, et al., Compromised melanocyte survival due to decreased suppression of CD4+ & CD8+ resident memory T cells by impaired TRM-regulatory T cells in generalized vitiligo patients, Exp. Dermatol. 33 (2024), e14982.
    [22]
    T. Czarnowicki, H. He, A. Leonard, et al., Blood endotyping distinguishes the profile of vitiligo from that of other inflammatory and autoimmune skin diseases, J. Allergy Clin. Immunol. 143 (2019) 2095-2107.
    [23]
    C.M. Mousset, W. Hobo, R. Woestenenk, et al., Comprehensive phenotyping of T cells using flow cytometry, Cytometry A. 95 (2019) 647-654.
    [24]
    E.J. Wherry, V. Teichgraber, T.C. Becker, et al., Lineage relationship and protective immunity of memory CD8 T cell subsets, Nat. Immunol. 4 (2003) 225-234.
    [25]
    J.J. Goronzy, C.M. Weyand, T cell development and receptor diversity during aging, Curr. Opin. Immunol. 17 (2005) 468-475.
    [26]
    V. Eleftheriadou, R. Atkar, J. Batchelor, et al., British Association of Dermatologists guidelines for the management of people with vitiligo 2021, Br. J. Dermatol. 186 (2022) 18-29.
    [27]
    N. van Geel, L. Depaepe, V. Vandaele, et al., Assessing the dynamic changes in vitiligo: reliability and validity of the Vitiligo Disease Activity Score (VDAS) and Vitiligo Disease Improvement Score (VDIS), J. Eur. Acad. Dermatol. Venereol. 36 (2022) 1334-1341.
    [28]
    N. van Geel, T. Passeron, A. Wolkerstorfer, et al., Reliability and validity of the Vitiligo Signs of Activity Score (VSAS), Br. J. Dermatol. 183 (2020) 883-890.
    [29]
    K. He, W. Wu, X. Wang, et al., Circulatory levels of alarmins in patients with non-segmental vitiligo: Potential biomarkers for disease diagnosis and activity/severity assessment, Front. Immunol. 13 (2022) 1069196.
    [30]
    S. Li, W. Dai, S. Wang, et al., Clinical significance of serum oxidative stress markers to assess disease activity and severity in patients with non-segmental vitiligo, Front. Cell. Dev. Biol. 9 (2021), 739413.
    [31]
    R. Speeckaert, J. Lambert, N. van Geel, Clinical significance of serum soluble CD molecules to assess disease activity in vitiligo, JAMA Dermatol. 152 (2016) 1194-1200.
    [32]
    J.P. Strassner, M. Rashighi, M. Ahmed Refat, et al., Suction blistering the lesional skin of vitiligo patients reveals useful biomarkers of disease activity, J. Am. Acad. Dermatol. 76 (2017) 847-855.e5.
    [33]
    X.X. Wang, Q.Q. Wang, J.Q. Wu, et al., Increased expression of CXCR3 and its ligands in patients with vitiligo and CXCL10 as a potential clinical marker for vitiligo, Br. J. Dermatol. 174 (2016) 1318-1326.
    [34]
    I. Odak, J. Barros-Martins, B. Bosnjak, et al., Reappearance of effector T cells is associated with recovery from COVID-19, EBioMedicine 57 (2020), 102885.
    [35]
    R. Kratchmarov, A.M. Magun, S.L. Reiner, TCF1 expression marks self-renewing human CD8+ T cells, Blood Adv. 2 (2018) 1685-1690.
    [36]
    M.C. van Aalderen, M. van den Biggelaar, E.B.M. Remmerswaal, et al., Label-free analysis of CD8+ T cell subset proteomes supports a progressive differentiation model of human-virus-specific T cells, Cell Rep. 19 (2017) 1068-1079.
    [37]
    P. Durek, K. Nordstrom, G. Gasparoni, et al., Epigenomic profiling of human CD4+ T cells supports a linear differentiation model and highlights molecular regulators of memory development, Immunity 45 (2016) 1148-1161.
    [38]
    J.G. van den Boorn, D. Konijnenberg, T.A. Dellemijn, et al., Autoimmune destruction of skin melanocytes by perilesional T cells from vitiligo patients, J. Invest. Dermatol. 129 (2009) 2220-2232.
    [39]
    B. Palermo, R. Campanelli, S. Garbelli, et al., Specific cytotoxic T lymphocyte responses against Melan-A/MART1, tyrosinase and gp100 in vitiligo by the use of major histocompatibility complex/peptide tetramers: The role of cellular immunity in the etiopathogenesis of vitiligo, J. Invest. Dermatol. 117 (2001) 326-332.
  • 加载中

Catalog

    通讯作者: 陈斌, bchen63@163.com
    • 1. 

      沈阳化工大学材料科学与工程学院 沈阳 110142

    1. 本站搜索
    2. 百度学术搜索
    3. 万方数据库搜索
    4. CNKI搜索

    Figures(1)

    Article Metrics

    Article views (52) PDF downloads(6) Cited by()
    Proportional views
    Related

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return