Mixed signature of activation and dysfunction allows human decidual CD8+ T cells to provide both tolerance and immunity

Anita van der Zwan, Kevin Bi, Errol R Norwitz, Ângela C Crespo, Frans H J Claas, Jack L Strominger, Tamara Tilburgs, Anita van der Zwan, Kevin Bi, Errol R Norwitz, Ângela C Crespo, Frans H J Claas, Jack L Strominger, Tamara Tilburgs

Abstract

Understanding how decidual CD8+ T cell (CD8+ dT) cytotoxicity is regulated and how these cells integrate the competing needs for maternal-fetal tolerance and immunity to infection is an important research and clinical goal. Gene-expression analysis of effector-memory CD8+ dT demonstrated a mixed transcriptional signature of T cell dysfunction, activation, and effector function. High protein expression of coinhibitory molecules PD1, CTLA4, and LAG3, accompanied by low expression of cytolytic molecules suggests that the decidual microenvironment reduces CD8+ dT effector responses to maintain tolerance to fetal antigens. However, CD8+ dT degranulated, proliferated, and produced IFN-γ, TNF-α, perforin, and granzymes upon in vitro stimulation, demonstrating that CD8+ dT are not permanently suppressed and retain the capacity to respond to proinflammatory events, such as infections. The balance between transient dysfunction of CD8+ dT that are permissive of placental and fetal development, and reversal of this dysfunctional state, is crucial in understanding the etiology of pregnancy complications and prevention of congenital infections.

Keywords: T cell exhaustion; cytotoxicity; placenta; pregnancy; trophoblast.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Fig. 1.
Fig. 1.
Transcriptional signatures of CD8+ EM dT dysfunction, activation, and effector function are coupled. (A) Unsupervised PCA analysis, using all 54,715 probes, of first trimester (dT 6–12 wk) and term pregnancy (dT > 37 wk) CD8+ EM dT and EM pT. (B) GSEA performed with assorted immunological signatures, showing positive correlation for an in vitro CD8+ activation signature, an effector signature, and a CD8+ viral exhaustion signature (MSigDB). (C) GSEA comparing first trimester CD8+ EM dT to gene modules of dysfunction, activation, and activation coupled to dysfunction (16).
Fig. 2.
Fig. 2.
Stimulation of CD8+ EM dT generates signatures of T cell activation. (A) A MaSigPro time-course analysis of ∼2,000 immunological relevant genes preselected based on the Immune System Process GO terms expressed by first trimester CD8+ EM dT stimulated with anti-CD3/CD28 in the presence of 50 U/mL IL-2 for 0, 12, or 72 h. Five clusters of temporally sensitive genes were identified. (B) K-Cluster analysis highlighting select genes in clusters 3–5.
Fig. 3.
Fig. 3.
CD8+ dT are functional upon activation. (A) FACS plots (Left) and percentages (Right) of CD107a+CD8+ pT and dT after stimulation with PMA/Ionomycin (1 μg/mL) for 6 h. Percentage CD107a+ cells are depicted within total CD8+ T cells and the four CD8+ T cell subpopulations (as percentage of total CD8). (B) Representative histograms (Left) and percentage divided cells (Right) of CFSE-labeled total CD8+ T cells at days 3, 4, 5, and 6 after stimulation with anti-CD3/28 (2 μL/mL) and 50 U/mL IL-2. (C) FACS plots (Left) and percentages (Right) of expression of intracellular IFN-γ, TNF-α, and IL-2 in total CD8+ pT and dT stimulated with PMA/Ionomycin (1 μg/mL) for 6 h. Bars represent the median with interquartile range; data are representative of three to six independent experiments; *P ≤ 0.05, **P ≤ 0.01.
Fig. 4.
Fig. 4.
Decidual CD8+CD28− T cells increase PRF and GZMB upon activation. Histograms of intracellular PRF (A) and GZMB (B) expression in CD8+CD28− pT and dT cultured for 6 d in the presence of IL-2 (50 U/mL), TNF-α (20 ng), IL-12 (20 ng), anti-CD3/28 (2 μL/mL), or a combination of anti-CD3/28 and IL-12. The vertical line signifies the PRF and GZMB mean fluorescence intensity (MFI) after addition of a nonactivating concentration of IL-2.
Fig. 5.
Fig. 5.
PRF, but not GZMB, is suppressed in HCMV-specific CD8+CD28− dT. Histograms and MFI of PRF (A and B) and GZMB (C and D) expression in tetramer-positive CD8+CD28− pT and dT, directly ex vivo and after in vitro expansion. Bars depict the median with interquartile range; *P ≤ 0.05, **P ≤ 0.01.

References

    1. Tilburgs T, Strominger JL. CD8+ effector T cells at the fetal-maternal interface, balancing fetal tolerance and antiviral immunity. Am J Reprod Immunol. 2013;69:395–407.
    1. Schwartz RH. T cell anergy. Annu Rev Immunol. 2003;21:305–334.
    1. Virgin HW, Wherry EJ, Ahmed R. Redefining chronic viral infection. Cell. 2009;138:30–50.
    1. Wherry EJ, et al. Molecular signature of CD8+ T cell exhaustion during chronic viral infection. Immunity. 2007;27:670–684.
    1. Zhang JY, et al. PD-1 up-regulation is correlated with HIV-specific memory CD8+ T-cell exhaustion in typical progressors but not in long-term nonprogressors. Blood. 2007;109:4671–4678.
    1. Ahmadzadeh M, et al. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood. 2009;114:1537–1544.
    1. Zajac AJ, et al. Viral immune evasion due to persistence of activated T cells without effector function. J Exp Med. 1998;188:2205–2213.
    1. Gallimore A, et al. Induction and exhaustion of lymphocytic choriomeningitis virus-specific cytotoxic T lymphocytes visualized using soluble tetrameric major histocompatibility complex class I-peptide complexes. J Exp Med. 1998;187:1383–1393.
    1. Johnson S, et al. Protective efficacy of individual CD8+ T cell specificities in chronic viral infection. J Immunol. 2015;194:1755–1762.
    1. Draenert R, et al. Immune selection for altered antigen processing leads to cytotoxic T lymphocyte escape in chronic HIV-1 infection. J Exp Med. 2004;199:905–915.
    1. Timm J, et al. CD8 epitope escape and reversion in acute HCV infection. J Exp Med. 2004;200:1593–1604.
    1. Vieira Braga FA, Hertoghs KM, van Lier RA, van Gisbergen KP. Molecular characterization of HCMV-specific immune responses: Parallels between CD8(+) T cells, CD4(+) T cells, and NK cells. Eur J Immunol. 2015;45:2433–2445.
    1. Wu X, et al. PD-1(+) CD8(+) T cells are exhausted in tumours and functional in draining lymph nodes of colorectal cancer patients. Br J Cancer. 2014;111:1391–1399.
    1. Wykes MN, Lewin SR. Immune checkpoint blockade in infectious diseases. Nat Rev Immunol. October 9, 2017 doi: 10.1038/nri.2017.112.
    1. Baumeister SH, Freeman GJ, Dranoff G, Sharpe AH. Coinhibitory pathways in immunotherapy for cancer. Annu Rev Immunol. 2016;34:539–573.
    1. Singer M, et al. A distinct gene module for dysfunction uncoupled from activation in tumor-infiltrating T cells. Cell. 2016;166:1500–1511.e9.
    1. Hamer DH. Metallothionein. Annu Rev Biochem. 1986;55:913–951.
    1. Scaife PJ, Bulmer JN, Robson SC, Innes BA, Searle RF. Effector activity of decidual CD8+ T lymphocytes in early human pregnancy. Biol Reprod. 2006;75:562–567.
    1. Tilburgs T, et al. Human HLA-G+ extravillous trophoblasts: Immune-activating cells that interact with decidual leukocytes. Proc Natl Acad Sci USA. 2015;112:7219–7224.
    1. Wang SC, et al. PD-1 and Tim-3 pathways are associated with regulatory CD8+ T-cell function in decidua and maintenance of normal pregnancy. Cell Death Dis. 2015;6:e1738.
    1. Taglauer ES, Trikhacheva AS, Slusser JG, Petroff MG. Expression and function of PDCD1 at the human maternal-fetal interface. Biol Reprod. 2008;79:562–569.
    1. Tilburgs T, et al. Human decidual tissue contains differentiated CD8+ effector-memory T cells with unique properties. J Immunol. 2010;185:4470–4477.
    1. King A, et al. Surface expression of HLA-C antigen by human extravillous trophoblast. Placenta. 2000;21:376–387.
    1. Holland OJ, et al. Minor histocompatibility antigens are expressed in syncytiotrophoblast and trophoblast debris: Implications for maternal alloreactivity to the fetus. Am J Pathol. 2012;180:256–266.
    1. Bulmer JN, Morrison L, Longfellow M, Ritson A, Pace D. Granulated lymphocytes in human endometrium: Histochemical and immunohistochemical studies. Hum Reprod. 1991;6:791–798.
    1. Constantin CM, et al. Normal establishment of virus-specific memory CD8 T cell pool following primary infection during pregnancy. J Immunol. 2007;179:4383–4389.
    1. Clark DR, et al. Perinatal Listeria monocytogenes susceptibility despite preconceptual priming and maintenance of pathogen-specific CD8(+) T cells during pregnancy. Cell Mol Immunol. 2014;11:595–605.
    1. Moldenhauer LM, et al. Cross-presentation of male seminal fluid antigens elicits T cell activation to initiate the female immune response to pregnancy. J Immunol. 2009;182:8080–8093.
    1. van Kampen CA, et al. Pregnancy can induce long-persisting primed CTLs specific for inherited paternal HLA antigens. Hum Immunol. 2001;62:201–207.
    1. Verdijk RM, et al. Pregnancy induces minor histocompatibility antigen-specific cytotoxic T cells: Implications for stem cell transplantation and immunotherapy. Blood. 2004;103:1961–1964.
    1. Meuleman T, et al. HLA-C antibodies in women with recurrent miscarriage suggests that antibody mediated rejection is one of the mechanisms leading to recurrent miscarriage. J Reprod Immunol. 2016;116:28–34.
    1. Lissauer D, et al. Cytomegalovirus sero positivity dramatically alters the maternal CD8+ T cell repertoire and leads to the accumulation of highly differentiated memory cells during human pregnancy. Hum Reprod. 2011;26:3355–3365.
    1. van Egmond A, van der Keur C, Swings GM, Scherjon SA, Claas FH. The possible role of virus-specific CD8(+) memory T cells in decidual tissue. J Reprod Immunol. 2016;113:1–8.
    1. Romero P, et al. Four functionally distinct populations of human effector-memory CD8+ T lymphocytes. J Immunol. 2007;178:4112–4119.
    1. Subramanian A, et al. Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 2005;102:15545–15550.
    1. Godec J, et al. Compendium of immune signatures identifies conserved and species-specific biology in response to inflammation. Immunity. 2016;44:194–206.
    1. Wu C, et al. Galectin-9-CD44 interaction enhances stability and function of adaptive regulatory T cells. Immunity. 2014;41:270–282.
    1. Paiva P, et al. Interleukin-11 promotes migration, but not proliferation, of human trophoblast cells, implying a role in placentation. Endocrinology. 2007;148:5566–5572.
    1. Crespo AC, Strominger JL, Tilburgs T. Expression of KIR2DS1 by decidual natural killer cells increases their ability to control placental HCMV infection. Proc Natl Acad Sci USA. 2016;113:15072–15077.
    1. Barton BM, Xu R, Wherry EJ, Porrett PM. Pregnancy promotes tolerance to future offspring by programming selective dysfunction in long-lived maternal T cells. J Leukoc Biol. 2017;101:975–987.
    1. Tilburgs T, Evans JH, Crespo AC, Strominger JL. The HLA-G cycle provides for both NK tolerance and immunity at the maternal-fetal interface. Proc Natl Acad Sci USA. 2015;112:13312–13317.
    1. Kopcow HD, et al. Human decidual NK cells form immature activating synapses and are not cytotoxic. Proc Natl Acad Sci USA. 2005;102:15563–15568.
    1. Siewiera J, et al. Human cytomegalovirus infection elicits new decidual natural killer cell effector functions. PLoS Pathog. 2013;9:e1003257, and erratum (2013) 9:10.1371.
    1. Trifari S, et al. MicroRNA-directed program of cytotoxic CD8+ T-cell differentiation. Proc Natl Acad Sci USA. 2013;110:18608–18613.

Source: PubMed

3
Subscribe