CCR4+ Skin-Tropic Phenotype as a Feature of Central Memory CD8+ T Cells in Healthy Subjects and Psoriasis Patients

Fabio Casciano, Marco Diani, Andrea Altomare, Francesca Granucci, Paola Secchiero, Giuseppe Banfi, Eva Reali, Fabio Casciano, Marco Diani, Andrea Altomare, Francesca Granucci, Paola Secchiero, Giuseppe Banfi, Eva Reali

Abstract

The chemokine receptor CCR4 has emerged as a skin-homing molecule important for the migration of T cells from the blood to the dermis. From our previous data on psoriasis patients, CCR4+ memory T cells emerged as a putative recirculating population between skin and blood. Here we focused our attention on the expression of CCR4 and skin-tropic molecules in the different stages of memory T cell differentiation. We analyzed the chemokine receptor profile in CD8+ and CD4+ CD45RA-CCR7+ (TCM) and CD45RA-CCR7- (TEM) cells. Subpopulations were further divided on the basis of CD62L expression, and the distribution among the subsets of the skin-homing molecule CLA (Cutaneous Lymphocyte Antigen) was evaluated. The characterization was performed on peripheral blood mononuclear cells isolated from 21 healthy subjects and 24 psoriasis patients. The results indicate that (i) the skin-homing CCR4 marker is mainly expressed in TCM cells, (ii) CCR4+ TCM cells also express high level of CLA and that (iii) the more differentiated phenotype TEM expresses CXCR3 and CCR5 but lower level of CCR4 and CLA. This indicates that progressive stages of memory T cell differentiation have profoundly different chemokine receptor patterns, with CD8+ TCM displaying a marked skin-tropic phenotype CLA+CCR4+. Differential skin-tropic phenotype between TCM and TEM cells was observed in both healthy subjects and psoriasis patients. However, patients showed an expanded circulating population of CD8+ TCM cells with phenotype CCR4+CXCR3+ that could play a role in the pathophysiology of psoriasis and possibly in disease recurrence.

Trial registration: ClinicalTrials.gov NCT03374527.

Keywords: T cells; central memory; effector memory; psoriatic disease; skin; tissue immunosurveillance.

Copyright © 2020 Casciano, Diani, Altomare, Granucci, Secchiero, Banfi and Reali.

Figures

Figure 1
Figure 1
Differential expression of CCR4 in CD8+ TCM and TEM cells. PBMCs isolated from healthy control subjects were stained for CD8, memory T cell phenotype markers (CD45RA and CCR7) and for chemokine receptors CCR4 and CCR5. (A) CD8+ T cells gated as CD45RA−CCR7+ TCM and CD45RA−CCR7− TEM were analyzed for the expression of CCR4 and CCR5. Representative analysis is shown in the figure. The axis scales for fluorescence are reported as log. Statistical analysis of the differences was performed by Mann–Whitney test. p-values < 0.05 were considered significant: ****p < 0.0001. (B) Mean values of the percentage of CCR4/CCR5 subpopulations among TCM and TEM cells were shown in pie charts.
Figure 2
Figure 2
CCR4 expression characterizes the TCM compartment. PBMCs isolated from healthy control subjects were stained for CD8, memory T cell phenotype markers (CD45RA and CCR7) and for chemokine receptors CCR4 and CCR5. (A) CD8+ T cells were analyzed for the memory phenotype according to CD45RA, CCR7 expression and for the expression of the chemokine receptors CCR4 and CCR5. On the basis of the chemokine receptor expression we identified five subsets CCR4hi (gate 1), CCR4int (gate 2), CCR4−CCR5− (gate 3), CCR4−CCR5+ (gate 4) and CCR4+CCR5+ (gate 5). These five subsets were superimposed to the density plot of the CD45RA− gated cells. Each red dot identifies cells from the corresponding subset as reported in the figure. The axis scales for fluorescence are reported as log.
Figure 3
Figure 3
CLA expression is maximal in CCR4+ cells and considerably decreases in CXCR3+ cells. PBMCs isolated from healthy control subjects were stained for CD8, memory T cell phenotype markers (CD45RA and CCR7), for chemokine receptors CCR4, CXCR3, and for CLA. (A) CD8+ T cells gated as TCM and TEM were analyzed for the expression of CCR4 and CXCR3. Statistical analysis of the differences was performed by Mann-Whitney test. p-values < 0.05 were considered significant: ***p < 0.001, ****p < 0.0001. Mean values for CCR4/CXCR3 subpopulations among TCM and TEM cells were shown in pie charts. (B) TCM and TEM cells were analyzed for differences in the percentage of CLA+ cells in the CCR4+CXCR3−, CCR4+CXCR3+, and CXCR3+ subpopulations using Mann-Whitney test. p-values < 0.05 were considered significant: ****p < 0.0001. (C) Reverse analysis is shown in the panel. Total memory CD45RA− CD8+ T cells were gated and analyzed for the expression of CLA and for chemokine receptors CCR4/CXCR3. On the basis of the chemokine receptor expression we identified five subsets CCR4hi (gate 1), CCR4int (gate 2), CCR4−CXCR3− (gate 3), CCR4−CXCR3+ (gate 4), CCR4+CXCR3+ (gate 5). The five subsets of CCR4/CXCR3 expressing cells were overlaid to the density plot of CD45RA− cells. Each red dot represents a cell from the corresponding subset. Among those dots, green dots identify cells of the gated subsets (CCR4hi, CCR4int, CCR4−CXCR3−, CCR4−CXCR3+, and CCR4+CXCR3+) that co-express CLA. The axis scales for fluorescence are reported as log.
Figure 4
Figure 4
CD62L+ CCR7+ central memory T cells have a skin-tropic phenotype. (A) Representative analysis of chemokine receptor expression in CD62L+/− gated CD45RA− CCR7+ (TCM) CD8+ T cells. Each subset (CCR4+CXCR3−, CCR4+CXCR3+, and CCR4−CXCR3+) in CD62L+ and CD62L− gates was also analyzed for the expression of CLA. The axis scales for fluorescence are reported as log and the axis scale for FSC is reported as linear. (B) CD45RA−CCR7+ (TCM) CD8+ T cells were gated on the basis of the expression of CD62L and analyzed for the expression of chemokine receptors (CCR4/CXCR3). The percentage of positive cells for each phenotype in CD62L+ and CD62L− gated cells was represented as Tukey's boxplot. Significance of the differences was calculated using Student's t-test for paired samples. p-values < 0.05 were considered significant: *p < 0.05. (C) CD45RA− CCR7+ (TCM) CD8+ T cells were gated on the basis of the expression of CD62L and further divided into different chemokine expressing subsets. The percentages of CLA+ cells in the different subsets are represented as Tukey's boxplot. Significance of the differences was calculated using Student's t-test for paired samples. p-values < 0.05 were considered significant: *p < 0.05.
Figure 5
Figure 5
Skin-tropic phenotype of circulating CD8+ T cells in psoriasis patients. (A) CD8+ cells from patients with cutaneous psoriasis (PsO) and healthy subjects were analyzed for the percentage of cells expressing chemokine receptors CCR4/CXCR3 in the CD45RA−CCR7+ (TCM) compartment. Statistical analysis of the differences was performed by Mann–Whitney test. p-values < 0.05 were considered significant: **p < 0.01. (B) CD8+ cells gated on the basis of the memory TCM or effector TEM phenotype, from patients with cutaneous psoriasis (PsO) and healthy subjects were analyzed for the percentage of CLA expressing cells in the CCR4+CXCR3− subpopulation. Statistical analysis of the differences was performed by Mann–Whitney test. p-values <0.05 were considered significant: *p < 0.05.

References

    1. Sallusto F, Lenig D, Forster R, Lipp M, Lanzavecchia A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature. (1999) 401:708–12. 10.1038/44385
    1. Mueller SN, Gebhardt T, Carbone FR, Heath WR. Memory T cell subsets, migration patterns, and tissue residence. Annu Rev Immunol. (2013) 31:137–61. 10.1146/annurev-immunol-032712-095954
    1. Tian Y, Babor M, Lane J, Schulten V, Patil VS, Seumois G, et al. . Unique phenotypes and clonal expansions of human CD4 effector memory T cells re-expressing CD45RA. Nat Commun. (2017) 8:1473. 10.1038/s41467-017-01728-5
    1. Mahnke YD, Brodie TM, Sallusto F, Roederer M, Lugli E. The who's who of T-cell differentiation: human memory T-cell subsets. Eur J Immunol. (2013) 43:2797–809. 10.1002/eji.201343751
    1. Restifo NP, Gattinoni L. Lineage relationship of effector and memory T cells. Curr Opin Immunol. (2013) 25:556–63. 10.1016/j.coi.2013.09.003
    1. Sallusto F, Langenkamp A, Geginat J, Lanzavecchia A. Functional subsets of memory T cells identified by CCR7 expression. Curr Top Microbiol Immunol. (2000) 251:167–71. 10.1007/978-3-642-57276-0_21
    1. Sallusto F, Geginat J, Lanzavecchia A. Central memory and effector memory T cell subsets: function, generation, and maintenance. Annu Rev Immunol. (2004) 22:745–63. 10.1146/annurev.immunol.22.012703.104702
    1. Sallusto F. Heterogeneity of human CD4(+) T cells against microbes. Annu Rev Immunol. (2016) 34:317–34. 10.1146/annurev-immunol-032414-112056
    1. Farber DL, Yudanin NA, Restifo NP. Human memory T cells: generation, compartmentalization and homeostasis. Nat Rev Immunol. (2014) 14:24–35. 10.1038/nri3567
    1. Heath WR, Carbone FR. The skin-resident and migratory immune system in steady state and memory: innate lymphocytes, dendritic cells and T cells. Nat Immunol. (2013) 14:978–85. 10.1038/ni.2680
    1. Ho AW, Kupper TS. T cells and the skin: from protective immunity to inflammatory skin disorders. Nat Rev Immunol. (2019) 19:490–502. 10.1038/s41577-019-0162-3
    1. Gebhardt T, Mueller SN, Heath WR, Carbone FR. Peripheral tissue surveillance and residency by memory T cells. Trends Immunol. (2013) 34:27–32. 10.1016/j.it.2012.08.008
    1. Gaide O, Emerson RO, Jiang X, Gulati N, Nizza S, Desmarais C, et al. . Common clonal origin of central and resident memory T cells following skin immunization. Nat Med. (2015) 21:647–53. 10.1038/nm.3860
    1. Thome JJ, Farber DL. Emerging concepts in tissue-resident T cells: lessons from humans. Trends Immunol. (2015) 36:428–35. 10.1016/j.it.2015.05.003
    1. Gehad A, Teague JE, Matos TR, Huang V, Yang C, Watanabe R, et al. . A primary role for human central memory cells in tissue immunosurveillance. Blood Adv. (2018) 2:292–8. 10.1182/bloodadvances.2017011346
    1. Park CO, Fu X, Jiang X, Pan Y, Teague JE, Collins N, et al. . Staged development of long-lived T-cell receptor alphabeta TH17 resident memory T-cell population to candida albicans after skin infection. J Allergy Clin Immunol. (2018) 142:647–62. 10.1016/j.jaci.2017.09.042
    1. Sallusto F, Cassotta A, Hoces D, Foglierini M, Lanzavecchia A. Do memory CD4 T cells keep their cell-type programming: plasticity versus fate commitment? T-cell heterogeneity, plasticity, and selection in humans. Cold Spring Harb Perspect Biol. (2018) 10:a029421. 10.1101/cshperspect.a029421
    1. Lugli E, Hudspeth K, Roberto A, Mavilio D. Tissue-resident and memory properties of human T-cell and NK-cell subsets. Eur J Immunol. (2016) 46:1809–17. 10.1002/eji.201545702
    1. Sgambelluri F, Diani M, Altomare A, Frigerio E, Drago L, Granucci F, et al. . A role for CCR5(+)CD4 T cells in cutaneous psoriasis and for CD103(+) CCR4(+) CD8 Teff cells in the associated systemic inflammation. J Autoimmun. (2016) 70:80–90. 10.1016/j.jaut.2016.03.019
    1. Voltan R, Rimondi E, Melloni E, Rigolin GM, Casciano F, Arcidiacono MV, et al. . Ibrutinib synergizes with MDM-2 inhibitors in promoting cytotoxicity in B chronic lymphocytic leukemia. Oncotarget. (2016) 7:70623–38. 10.18632/oncotarget.12139
    1. Baumgarth N, Roederer M. A practical approach to multicolor flow cytometry for immunophenotyping. J Immunol Methods. (2000) 243:77–97. 10.1016/S0022-1759(00)00229-5
    1. Roederer M. Spectral compensation for flow cytometry: visualization artifacts, limitations, and caveats. Cytometry. (2001) 45:194–205. 10.1002/1097-0320(20011101)45:3<194::AID-CYTO1163>;2-C
    1. Maecker HT, Trotter J. Flow cytometry controls, instrument setup, and the determination of positivity. Cytometry A. (2006) 69:1037–42. 10.1002/cyto.a.20333
    1. Diani M, Galasso M, Cozzi C, Sgambelluri F, Altomare A, Cigni C, et al. . Blood to skin recirculation of CD4(+) memory T cells associates with cutaneous and systemic manifestations of psoriatic disease. Clin Immunol. (2017) 180:84–94. 10.1016/j.clim.2017.04.001
    1. Ma CS, Wong N, Rao G, Nguyen A, Avery DT, Payne K, et al. . Unique and shared signaling pathways cooperate to regulate the differentiation of human CD4+ T cells into distinct effector subsets. J Exp Med. (2016) 213:1589–608. 10.1084/jem.20151467
    1. Watanabe R, Gehad A, Yang C, Scott LL, Teague JE, Schlapbach C, et al. . Human skin is protected by four functionally and phenotypically discrete populations of resident and recirculating memory T cells. Sci Transl Med. (2015) 7:279ra239. 10.1126/scitranslmed.3010302
    1. Kunkel EJ, Boisvert J, Murphy K, Vierra MA, Genovese MC, Wardlaw AJ, et al. . Expression of the chemokine receptors CCR4, CCR5, and CXCR3 by human tissue-infiltrating lymphocytes. Am J Pathol. (2002) 160:347–55. 10.1016/S0002-9440(10)64378-7
    1. Geginat J, Lanzavecchia A, Sallusto F. Proliferation and differentiation potential of human CD8+ memory T-cell subsets in response to antigen or homeostatic cytokines. Blood. (2003) 101:4260–6. 10.1182/blood-2002-11-3577
    1. Campbell JJ, Clark RA, Watanabe R, Kupper TS. Sezary syndrome and mycosis fungoides arise from distinct T-cell subsets: a biologic rationale for their distinct clinical behaviors. Blood. (2010) 116:767–71. 10.1182/blood-2009-11-251926
    1. Tubo NJ, McLachlan JB, Campbell JJ. Chemokine receptor requirements for epidermal T-cell trafficking. Am J Pathol. (2011) 178:2496–503. 10.1016/j.ajpath.2011.02.031
    1. Clark RA, Chong B, Mirchandani N, Brinster NK, Yamanaka K, Dowgiert RK, et al. . The vast majority of CLA+ T cells are resident in normal skin. J Immunol. (2006) 176:4431–9. 10.4049/jimmunol.176.7.4431
    1. Campbell JJ, O'Connell DJ, Wurbel MA. Cutting edge: chemokine receptor CCR4 is necessary for antigen-driven cutaneous accumulation of CD4 T cells under physiological conditions. J Immunol. (2007) 178:3358–62. 10.4049/jimmunol.178.6.3358
    1. Casciano F, Pigatto PD, Secchiero P, Gambari R, Reali E. T cell hierarchy in the pathogenesis of psoriasis and associated cardiovascular comorbidities. Front Immunol. (2018) 9:1390. 10.3389/fimmu.2018.01390
    1. Sells RE, Hwang ST. Paradoxical increase in skin inflammation in the absence of CCR4. J Invest Dermatol. (2010) 130:2697–9. 10.1038/jid.2010.292
    1. Gehad A, Al-Banna NA, Vaci M, Issekutz AC, Mohan K, Latta M, et al. . Differing requirements for CCR4, E-selectin, and alpha4beta1 for the migration of memory CD4 and activated T cells to dermal inflammation. J Immunol. (2012) 189:337–46. 10.4049/jimmunol.1102315
    1. Egawa G, Kabashima K. Skin as a peripheral lymphoid organ: revisiting the concept of skin-associated lymphoid tissues. J Invest Dermatol. (2011) 131:2178–85. 10.1038/jid.2011.198

Source: PubMed

3
Subscribe