CD8αα+T cells exert a pro-inflammatory role in patients with psoriasis

Y Y Zhang, Y T Lin, L Wang, X W Sun, E L Dang, K Xue, W G Zhang, K M Zhang, G Wang, B Li, Y Y Zhang, Y T Lin, L Wang, X W Sun, E L Dang, K Xue, W G Zhang, K M Zhang, G Wang, B Li

Abstract

Background: Psoriasis is a common chronic inflammatory disease caused by excessive activation of CD4+T cells, including Th17, Th1 and Th22. The role of CD8+T cells in psoriasis pathogenesis remains poorly understood.

Aim: To identify the phenotype of CD8+T cells in patients with psoriasis and to investigate its role in the formation of lesions.

Methods: The phenotype of CD8+T cells in psoriatic lesions was detected by immunofluorescence staining. Flow cytometry was performed to detect their phenotype in peripheral blood. Thereafter, coculture of CD8αα+T cells with autogenous CD4+T cells was performed to investigate the function of CD8αα+T cells in patients with psoriasis. Finally, pro-inflammatory factors produced by CD8αα+T cells were examined by immunofluorescence staining and flow cytometry.

Results: Compared to the CD8αβ+T cells, CD8αα+T cell infiltration in psoriatic lesions markedly increased. Moreover, epidermal CD8αα+T cells exhibited tissue-resident memory T cells (TRM) phenotypes and dermal CD8αα+T cells exhibited effector memory (TEM) phenotypes in psoriatic lesions. Additionally, we found that CD8αα+T cells from patients with psoriasis did not express the markers of regulatory T cells and could promote the proliferation of CD4+T effector cells and produce interleukin-17 and interferon-γ.

Conclusions: Our findings demonstrate that CD8αα+T cells contribute to the pathogenesis of psoriasis by producing pro-inflammatory factors.

Conflict of interest statement

The authors have no potential conflict of interest to declare.

© 2021 The Authors. Skin Health and Disease published by John Wiley & Sons Ltd on behalf of British Association of Dermatologists.

Figures

FIGURE 1
FIGURE 1
(a–f) CD8αα+T cells increasingly infiltrate into psoriatic lesions. Representative immunofluorescence staining of CD8α co‐expressed with (a) TCRα, TCRδ and (b) CD8β in psoriatic lesions and normal skin. (c) Proportion of CD8αβ+T and CD8αα+T cells determined by analysis of immunofluorescence staining (n = 8). (d and e) Frequency of circulating CD8αβ+T and CD8αα+T cells (bottom) in gated CD3+CD8+T cells (up) from patients with psoriasis (n = 32) and healthy controls (n = 16). (f) Frequency correlation of circulating CD8αα+T cells with psoriasis area and severity index (PASI) scores of patients with psoriasis (n = 32). **p < 0.01
FIGURE 2
FIGURE 2
(a–d) Epidermal CD8αα+T cells exhibit TRM phenotypes. Representative immunofluorescence staining of (a) CD103 and (c) CD69 expressed in CD8αα+T cells of psoriatic lesions and normal skin. Proportion of (b) CD103+CD8+TRM and (d) CD69+CD8+TRM cells determined by analysis of immunofluorescence staining (n = 8). **p < 0.01
FIGURE 3
FIGURE 3
CD8αα+T cells do not express markers of Treg cells in psoriatic lesions. Representative immunofluorescence staining of Foxp3, CD25 and CD122 expressed in CD8αα+T cells of psoriatic lesions and normal skin
FIGURE 4
FIGURE 4
(a–c) CD8αα+T cells exhibit a pro‐inflammatory role in patients with psoriasis. (a) CD8αα+T cells from patients with psoriasis or healthy controls were cocultured with autogenetic CD4+T cells. Proliferation of CD4+T cells was detected by flow cytometry, which could reflect the function of CD8αα+T cells. (b) Statistical analysis of flow cytometry. (c) Mean difference of CD4+T cell proliferation (proportion of proliferated CD4+T cells in the coculture group minus that in the group of autogenetic CD4+T cells alone, n = 5). **p < 0.01
FIGURE 5
FIGURE 5
(a–f) CD8αα+T cells produce pro‐inflammatory factors in patients with psoriasis. (a) Representative immunofluorescence staining of IL‐17A and interferon‐γ (IFN‐γ) expressed in CD8αα+T cells of psoriatic lesions and normal skin. (b) Proportion of IL‐17A+ CD8αα+T and IFN‐γ+CD8αα+T cells determined by analysis of immunofluorescence staining (n = 8). (c) The expression of IL‐17A and (d) IFN‐γ in circulating CD8αα+T cells from patients with psoriasis (n = 32) and healthy controls (n = 16) by flow cytometry. (e and f) Statistical analysis of flow cytometry. *p < 0.05; **p < 0.01; ***p < 0.001

References

    1. Lowes MA, Suarez‐Farinas M, Krueger JG. Immunology of psoriasis. Annu Rev Immunol. 2014;32:227–55.
    1. Boehncke WH, Schon MP. Psoriasis. Lancet. 2015;386:983–94.
    1. Hijnen D, Knol EF, Gent YY, Giovannone B, Beijn SJ, Kupper TS, et al. CD8(+) T cells in the lesional skin of atopic dermatitis and psoriasis patients are an important source of IFN‐gamma, IL‐13, IL‐17, and IL‐22. J Invest Dermatol. 2013;133:973–9.
    1. Di Cesare A, Di Meglio P, Nestle FO. The IL‐23/Th17 axis in the immunopathogenesis of psoriasis. J Invest Dermatol. 2009;129:1339–50.
    1. Jeschke JC, Williams CB. Editorial: crossing the divide: a novel Cd8 enhancer with activity in CTLs and CD8alphaalpha+ dendritic cells. J Leukoc Biol. 2015;97:623–5.
    1. Wu J, Zhou C, Robertson J, Weng CC, Meistrich ML, Tailor RC, et al. Identification of a bone marrow‐derived CD8alphaalpha+ dendritic cell‐like population in inflamed autoimmune target tissue with capability of inducing T cell apoptosis. J Leukoc Biol. 2010;88:849–61.
    1. Cheroutre H, Lambolez F. Doubting the TCR coreceptor function of CD8alphaalpha. Immunity. 2008;28:149–59.
    1. Rybakin V, Clamme JP, Ampudia J, Yachi PP, Gascoigne NR. CD8alphaalpha and ‐alphabeta isotypes are equally recruited to the immunological synapse through their ability to bind to MHC class I. EMBO Rep. 2011;12:1251–6.
    1. Fanchiang SS, Cojocaru R, Othman M, Khanna R, Brooks MJ, Smith T, et al. Global expression profiling of peripheral Qa‐1‐restricted CD8alphaalpha+TCRalphabeta+ regulatory T cells reveals innate‐like features: implications for immune‐regulatory repertoire. Hum Immunol. 2012;73:214–22.
    1. Qiu Y, Peng K, Liu M, Xiao W, Yang H. CD8alphaalpha TCRalphabeta intraepithelial lymphocytes in the mouse gut. Dig Dis Sci. 2016;61:1451–60.
    1. Olivares‐Villagomez D, Van Kaer L. TL and CD8alphaalpha: enigmatic partners in mucosal immunity. Immunol Lett. 2010;134:1–6.
    1. Ruscher R, Hogquist KA. Development, ontogeny, and maintenance of TCRalphabeta(+) CD8alphaalpha IEL. Curr Opin Immunol. 2019;58:83–8.
    1. Di Meglio P, Villanova F, Navarini AA, Mylonas A, Tosi I, Nestle FO, et al. Targeting CD8(+) T cells prevents psoriasis development. J Allergy Clin Immunol. 2016;138:274–6.
    1. Ovigne JM, Baker BS, Brown DW, Powles AV, Fry L. Epidermal CD8+ T cells in chronic plaque psoriasis are Tc1 cells producing heterogeneous levels of interferon‐gamma. Exp Dermatol. 2001;10:168–74.
    1. Bai L, Peng H. Generating CD8alphaalpha IELs from two sources of thymic precursors. Cell Mol Immunol. 2018;15:640–1.
    1. Zhu J, Peng T, Johnston C, Phasouk K, Kask AS, Klock A, et al. Immune surveillance by CD8alphaalpha+ skin‐resident T cells in human herpes virus infection. Nature. 2013;497:494–7.
    1. Holler PD, Yamagata T, Jiang W, Feuerer M, Benoist C, Mathis D. The same genomic region conditions clonal deletion and clonal deviation to the CD8alphaalpha and regulatory T cell lineages in NOD versus C57BL/6 mice. Proc Natl Acad Sci U S A. 2007;104:7187–92.
    1. Poussier P, Ning T, Banerjee D, Julius M. A unique subset of self‐specific intraintestinal T cells maintains gut integrity. J Exp Med. 2002;195:1491–7.
    1. Tang X, Maricic I, Purohit N, Bakamjian B, Reed‐Loisel LM, Beeston T, et al. Regulation of immunity by a novel population of Qa‐1‐restricted CD8alphaalpha+TCRalphabeta+ T cells. J Immunol. 2006;177:7645–55.
    1. Sheng H, Marrero I, Maricic I, Fanchiang SS, Zhang S, Sant'Angelo DB, et al. Distinct PLZF(+)CD8alphaalpha(+) Unconventional T Cells Enriched in Liver Use a Cytotoxic Mechanism to Limit Autoimmunity. J Immunol. 2019;203:2150–62.
    1. Sasson SC, Gordon CL, Christo SN, Klenerman P, Mackay LK. Local heroes or villains: tissue‐resident memory T cells in human health and disease. Cell Mol Immunol. 2020;17:113–22.
    1. Wang X, Tian Z, Peng H. Tissue‐resident memory‐like ILCs: innate counterparts of TRM cells. Protein Cell. 2020;11:85–96.
    1. Dijkgraaf FE, Matos TR, Hoogenboezem M, Toebes M, Vredevoogd DW, Mertz M, et al. Tissue patrol by resident memory CD8(+) T cells in human skin. Nat Immunol. 2019;20:756–64.
    1. Vo S, Watanabe R, Koguchi‐Yoshioka H, Matsumura Y, Ishitsuka Y, Nakamura Y, et al. CD8 resident memory T cells with interleukin 17A‐producing potential are accumulated in disease‐naive nonlesional sites of psoriasis possibly in correlation with disease duration. Br J Dermatol. 2019;181:410–2.
    1. Kurihara K, Fujiyama T, Phadungsaksawasdi P, Ito T, Tokura Y. Significance of IL‐17A‐producing CD8(+)CD103(+) skin resident memory T cells in psoriasis lesion and their possible relationship to clinical course. J Dermatol Sci. 2019;95:21–7.
    1. Cheuk S, Schlums H, Gallais Serezal I, Martini E, Chiang SC, Marquardt N, et al. CD49a expression defines tissue‐resident CD8(+) T cells poised for cytotoxic function in human skin. Immunity. 2017;46:287–300.

Source: PubMed

3
Sottoscrivi