uva-dare (digital academic repository) t cell development in … · infected individuals, the...

19
UvA-DARE is a service provided by the library of the University of Amsterdam (https://dare.uva.nl) UvA-DARE (Digital Academic Repository) T cell development in human cytomegalovirus infection Gamadia, L.E. Publication date 2003 Link to publication Citation for published version (APA): Gamadia, L. E. (2003). T cell development in human cytomegalovirus infection. General rights It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), other than for strictly personal, individual use, unless the work is under an open content license (like Creative Commons). Disclaimer/Complaints regulations If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You will be contacted as soon as possible. Download date:17 Jun 2021

Upload: others

Post on 01-Feb-2021

1 views

Category:

Documents


0 download

TRANSCRIPT

  • UvA-DARE is a service provided by the library of the University of Amsterdam (https://dare.uva.nl)

    UvA-DARE (Digital Academic Repository)

    T cell development in human cytomegalovirus infection

    Gamadia, L.E.

    Publication date2003

    Link to publication

    Citation for published version (APA):Gamadia, L. E. (2003). T cell development in human cytomegalovirus infection.

    General rightsIt is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s)and/or copyright holder(s), other than for strictly personal, individual use, unless the work is under an opencontent license (like Creative Commons).

    Disclaimer/Complaints regulationsIf you believe that digital publication of certain material infringes any of your rights or (privacy) interests, pleaselet the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the materialinaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letterto: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. Youwill be contacted as soon as possible.

    Download date:17 Jun 2021

    https://dare.uva.nl/personal/pure/en/publications/t-cell-development-in-human-cytomegalovirus-infection(656b01de-c22b-4da1-8c40-0b3a07768a72).html

  • C H A P T E R 5 THE SIZE AND PHENOTYPE OF VIRUS-

    S P E C I F I C T CELL POPULATIONS IS

    DETERMINED BY REPETITIVE

    ANTIGENIC STIMULATION AND

    ENVIRONMENTAL CYTOKINES

    Laila E. Gamadia1,2, Ester M.M. van Leeuwen1'2,

    Ester B.M. Remmerswaal2, Si-La Yong2,

    Sugianto Surachno', Pauline M.E. Wertheim-van

    Dillen3, Ineke J.M. ten Berge1'4 and René A.W.

    van Lier2.

    'Renal Transplant Unit, Department of Internal

    Medicine, laboratory for Experimental

    Immunology, 'Department of Virology and

    Division of Clinical Immunology and

    Rheumatology, Department of Internal Medicine,

    Academic Medical Centre, University of

    Amsterdam, Amsterdam, the Netherlands.

    Submitted

  • ABSTRACT

    Based on the expression of the TNF-R SFP CD27 two antigen-primed CD8 T cell subsets

    can be discerned in the circulation of healthy individuals: CD27~ T cells that secrete a variety

    of cytokines but do not display immediate cytolytic activity and CD27", cytotoxic T cells with

    limited cytokine production potential. The mechanism that controls the generation of these

    different phenotypes is unknown. We show that CMV reactivation not only increases the

    number of virus-specific T cells but induces their transition from a CD27' to a CD27"

    phcnotype. In support of a relation between pool-size and phenotype in a cohort of latently

    infected individuals, the number of antigen-specific CD27" CD8+ T cells was found to be

    linearly related to the total number of CMV specific CD8+ T cells. In vitro studies revealed

    that the acquisition of the CD27" phenotype on CMV-specific T cells depended on the

    interaction of CD27 with its cellular ligand, CD70. Expression of CD70 was proportional to

    the amount of antigenic stimulation and blocked by the CD4 T cell derived cytokine IL-21.

    Thus, induction of CD70, which may vary in distinct viral infections, appears to be a key

    factor in determining the size and phenotype of the CMV-specific T cell population in latently

    infected individuals.

    48

  • INTRODUCTION

    T cell memory is established after clearance of acute infection and maintained lifelong. The

    generation of CD8~ memory T cells is supposedly by the survival of a number of antigen-

    stimulated effector T cells that are generated early in the primary adaptive immune response

    to the causative pathogen. What determines a primary induced CD8+ T lymphocyte to survive

    into a long-lived memory cell is as yet unclear1. Primed T cells in the peripheral blood of

    healthy individuals show considerable phenotypic and functional heterogeneity. At one side

    of the spectrum are CD45R(TCD28+CD27+ T cells that produce a large variety of cytokines,

    but are unable to execute cytolysis without prior in vitro culture2. These cells have been

    defined as "memory-type" T cells because of their ability to clonally expand in response to

    recall antigens in the absence of helper T cell-derived cytokines. Based on the expression of

    the chemokine receptor CCR7, this population has been subdivided by Sallusto et al. into

    CCR7T "central-memory" T cells that have the ability to migrate to secondary lymphoid

    organs and CCR7" "effector-memory" T cells that do not have this ability. Although it was

    originally reported that CCR7+ and CCR7" T cells differ in immediate effector functions, this

    has been disputed by others4:5. Distinct from these "memory-type" cells is the population of

    CD45RA+CCR7-CD28-CD27" T cells (further referred to as CD45RA+CD27" T cells). Based

    on the expression of CD95-ligand, perforin and granzyme B and the ability to execute

    cytotoxicity directly ex vivo these T cells have been designated "effector-type" T cells. This

    population, that is also CD 11a'8 and CD57pos increases with age and has a restricted

    diversity of TCR-Vp genes6"8.

    T cells with specificities for different persistent viruses have been reported to vary in

    phenotype and function. For instance, EBV-specific T cells are predominantly

    CD45R(TCD28'CD27+ and in asymptomatic HIV-carriers, HIV-specific T cells are mostly

    CD45R0+CD28"CD27*, contrasting the phenotype of CMV-reactive T cells that are

    predominantly CD45RA"CD27"9. From these findings it was postulated that viruses might

    induce phenotypically distinct T cells . However, it is clear that the selection for phenotypes

    is not absolute since e.g. in healthy CMV carriers during latency virus-specific T cells with

    either CD45R0+CD27^ and CD45RA'CD27" surface profiles can be found 9: 10. Analysis of

    virus-specific T cells during acute infections with EBV, HIV or CMV revealed that early

    effector T cells are clearly distinct from the "effector-type" CD45RACD27" T cells found in

    49

  • latently infected persons. T cells expanding early in immune responses have CD45R0, CD28,

    CD27 surface expression, express the Gl phase-associated nuclear antigen K.i-67 and contain

    perforin and granzyme B " . These observations raised questions about the generation of

    CD45RA~CD27" T cells and their role in protective immunity9.

    Although it was originally proposed that CD45RAXD28CD27" T cells had entered a state of

    senescence , telomere length analyses revealed similar replicative histories in vivo of

    CD45R(fCD27T and CD45RACD27" CD8' T cells7. Moreover, it was shown that

    CD45RA CD27" T cells were unable to divide when stimulatory monoclonal antibodies to

    CD3 and CD28 are combined to activate these cells". Still, CMV-specific T cells with this

    phenotype potently expanded when stimulated by specific MHC/peptide ligands in the

    presence of helper T cell derived cytokinesl5; '6. These latter experiments implied that

    although CD45RA+CD27~ T cells are fully differentiated T cells where it concerns CTL

    effector functions they are not terminally differentiated in the sense of having lost the ability

    to yield progeny.

    Here, we studied patients with well-documented CMV reactivation to investigate the

    generation of CD27" T cells in vivo and their role in maintaining CMV latency. We show that

    "effector-type" T cells can expand in vivo when viral replication occurs. Moreover, antigenic

    load and helper T cell derived cytokines are key regulators of the phenotype of virus-specific

    T cells during latency, likely through regulation of the CD27 ligand, CD70.

    MATERIALS AND METHODS

    5 HLA-A2 positive and 1 HLA-B7 positive CMV-seropositive renal transplant recipients of

    either a kidney of a CMV-seropositive donor (2) or a CMV-seronegative donor (4) were

    longitudinally studied. Basic immunosuppressive therapy consisted of cyclosporin A, blood

    trough level aimed at 150 ng/mL. mycophenolate mofetil (lOOOmgs twice daily) and

    prednisone 10 mgs daily. Viral replication was monitored longitudinally by quantitative PCR

    from the timepoint of transplantation, and reactivation was determined by detectable viral

    loads above the cut-off point of 80 copies/mL. All subjects experienced asymptomatic CMV-

    reactivation and no anti-viral treatment was given. Heparinized peripheral blood samples were

    collected before transplantation and Meekly during 17 weeks after transplantation, whereafter

    samples were collected once a month. PBMC were isolated using standard density gradient

    50

  • centrifugation techniques and subsequently cryopreserved. In addition CMV-specific T cells

    were characterised in 11 healthy individuals. All patients gave written informed consent and

    the local medical ethical committee approved the study.

    Peptides

    The HLA-A2 binding CMV pp65 derived peptide NLVPMVATV and the HLA-B7 binding

    CMV pp65 derived peptide TPRVTGGGAM were purchased from the IHB-LUMC peptide

    synthesis library facility (Leiden, The Netherlands).

    Generation of tetrameric complexes

    Tetrameric complexes were manufactured at the tetramer facility of Sanquin, and generated

    essentially as described by Altman et al.18. In brief, purified HLA-A2.1 heavy chain or HLA-

    B7.2 heavy chain and p2 micro-globulin were synthesized using a prokaryotic expression

    system (pET; Novagen, Milwaukee, WI, USA). The heavy chain was modified by deletion of

    the transmembrane/cytosolic tail and COOH-terminal addition of a sequence containing the

    BirA enzymatic biotinylation site. The HLA-A2.1 binding CMV pp65 derived peptide

    NLVPMVATV and the HLA-B7.2 derived peptide TPRVTGGGAM were used for refolding.

    Monomeric complexes were concentrated, biotinylated by BirA (expressed using the pET

    expression system, purified using Clontech cobalt beads) in the presence of biotin (Molecular

    Probes), ATP (Sigma Chemical) and MgCl2. The biotinylated product was separated from

    free biotin by FPLC using a Superdex 200 ITR16 / 60 column (Amcrsham Pharmacia, Little

    Chalfont, UK). Streptavidin-allophycocyanin (APC) conjugate (Molecular Probes) was added

    in a 1:4 molar ratio and subsequently tetramers were FPLC purified using the same column.

    CFSE labelling

    PBMCs were pelleted and resuspended in PBS at a final concentration of 5-10 x 106 cells/ml.

    PBMCs were labeled with 2.5 |iM (final concentration) of 5-(and-6)-Carboxyfluorescein

    Diacetate Succinimidyl Ester (CFSE; Molecular Probes) in PBS for 8 minutes shaking at

    37°C. Cells were washed and subsequently resuspended in IMDM supplemented with 10%

    Human Pool Serum (HPS), antibiotics and 3.57 x 10"*% (v/v) (3-mercapto-ethanol (Merck)

    (culture medium).

    51

  • Culture and stimulation of cells

    CFSE labeled cells were cultured in culture medium for five days in 24 wells plates at a

    concentration of 0.5-1 x 106 cells/ml. CMVpp65-derived peptide was added at a final

    concentration of 1.25 |ig/ml. CMV-antigen (inactivated whole virus, 10 jil/'ml; Microbix) was

    used to stimulate cells. Furthermore, for stimulation IL-2 (50 U/ml; Biotest Ag, Dreieich,

    Germany), IL-15 (3 ng/ml; R&D systems, Abingdon, United Kingdom), IL-21 (50 ng/ml;

    Zymogenetics, Seattle, WA), were added. For blocking experiments we used CD70 antibody

    (clone2F2) at concentrations of 40(i.gr/mL. For dose dependent analysis of CD70 expression,

    1.25 jig peptide and 10 |iL CMV-antigen/ mL were titrated. Flow cytometric analysis was

    performed before culture and after five days.

    Immunofluorescent staining and flowcytometry of CMV-specific CD8+ T cells

    Thawed PBMC were resuspended in RPMI, containing 10% FCS and antibiotics. 200.000

    PBMC were incubated with fluorescent label conjugated mAbs (concentrations according to

    manufacturer's instructions) and an appropriate concentration of tetrameric complexes.

    Negative controls to validate specificity of the CMV-peptidc-tetrameric complexes consisted

    of HLA-A2.1/HLAB7.2 negative CMV-seropositive or HLA-A2.1/HLA-B7.2 positive CMV-

    seronegative healthy individuals and renal transplant recipients. Negative controls always

    showed tetramer staining of less than 0.01% of total lymphocytes (data not shown). For

    staining with the mouse anti-human CCR7 monoclonal antibody (mAb), a three step staining

    protocol was performed consisting of incubation with the CCR7 antibody (Pharmingen), for

    30 minutes, washing, incubation with biotinylated goat anti mouse IgM (Pharmingen) for 30

    minutes, incubation with 10% (v/v) normal mouse serum (Sanquin, Amsterdam, The

    Netherlands) followed by incubation with streptavidin-PE and directly conjugated mAbs and

    tetrameric complexes for 30 minutes. For staining with the mouse anti-human monoclonal

    antibody CD70 (clone 2F2) a two step staining protocol was performed consisting of

    incubation with the CD70 antibody for 30 minutes, washing, incubation with FITC

    conjugated anti-mouse IgGl antibodies, washing, incubation with 10% (v/v) normal mouse

    serum followed by incubation of directly conjugated mAbs. Analyses consisted of APC-

    conjugated tetramers and CD8-PerCP (BectonDickinson, San Jose, CA) in combination with

    either CD28 (Sanquin, the Netherlands) and CD27 (BD), CCR7 and CD45RA, CD27 and

    52

  • CD45RA (BD) and CD45RA and CD45R0 (BD), CD38 (BD) and CD70 all combinations in

    FITC and PE, and additional stainings for CD70 expression were done with CD70 in

    combination with CD4-PE, CD8-APC and CD19-PercP.

    CMV-PCR

    Quantitative PCR was performed in EDTA whole blood samples as described for plasma or 19

    serum .

    Statistical analyses

    Linear regression analysis using a mixed model was performed for the absolute numbers of

    total CMV-specific CD8+ T cells in relation with total CD27"CMV-specific CD8+ T cells, and

    for the percentage CMV-specific CD8+ T cells and the percentage CD27" of CMV-specific

    CD8+ T cells taken as a logit. Subsequently, the regression coefficient was calculated and

    tested for significance. Repeated observations were analysed by paired Students t-test or a

    repeated measures Anova, p-values, < 0.05 were considered statistically significant.

    RESULTS

    CMV-reactivation induces quantitative and qualitative changes in the CD8+ virus-

    specific T cell compartment

    In healthy individuals and renal transplant recipients during latency, CMV-specific CD8+ T

    cells may have different predominant phenotypes ranging from CD45R0+CD28+CD27+

    ("memory-type", patient 1, figure la, t=0) to CD45RAXD45R0duU'"CD27" ("effector-type"

    patient 2, figure Id, t=0)9: 1(). The fate of these distinct CMV-specific T cell populations was

    followed during CMV reactivation. In the patients with a CD45RA"CD28fCD27+ CD8^ T cell

    phenotype prior to reactivation a substantial number of these cells lost expression of CD28

    and CD27 (Fig 1, row a). Longitudinal analysis of CMV-specific CD8+CD27" T cells in these

    individuals showed that this subset increased immediately when replication of virus occurs,

    (Fig. 2a: CD27"tetramer' T cells in relation to viral load in one representative individual,

    Figure 2 b: CD27" tetramer+ T cells in all individuals; median before reactivation 26.5%,

    range 0.0 to 91.1 % median at the timepoint of reactivation 45.22%), and keeps on doing so

    long after viral replication has ceased (median at the first timepoint after reactivation 66.35 %,

    53

  • median at the last time-point measured, > 6 months after reactivation, 72.73%, p = 0.039). In

    the individuals with a predominant effector. CD45RA"CD28"CD27" CD8^ T cell phenotype,

    there was an increase in the number of CMV-specific T cells but no phenotypic change of

    virus-specific CD8' T cells with regard to the expression of CD28 and CD27 (Fig 1, row d).

    Patient 1 Patient 2

    rr>4SRft

    CD38 — •

    Figure 1: Quantitative and qualitative changes of CMV-specific CD8* memory cells during reactivation Figure la-c: Differentiation of CMV-specific CD8' T cells in one patient with a starting population of CD45R(fCD27XD28* CMV-specific cells. Figure ld-f: Differentiation of CMV-specific CD8* T cells in one patient with a starting population of CD45RA CD27CD28" cells. Time defined as t=0. before reactivation. t=l. during reactivation at peak PCR value and t=2. first timepoint after reactivation. All plots gated on CD8' T cells. CMV-specific CD8* T cells as defined by specific tetramer staining plotted in black, total CD84" T cells plotted in gray. Figure la and d: x-axis log fluorescence CD27-Fitc. y-axis log fluorescence CD28-PE; figure lb and e: x-axis log fluorescence CD45R0-PE, y-axis log fluorescence CD45RA-Fitc; figure lc and f: x-axis log fluorescence 38-PE, y-axis log fluorescence HLA-DR-Fitc.

    Irrespective of the pre-reactivation phenotype, the activation markers CD38 and HLA-DR

    were upregulated on CMV-specific cells in all patients at the time of viral replication (Fig. 1,

    rows c and f), as was the expression of CD45R0, reflecting recent proliferation of these cells

    (Fig. 1 rows b and e). CCR7 expression, documented to be upregulated upon activation161

    did not change in any of the patients during reactivation (data not shown), although re-

    expression of this marker could redirect subjected cells to secondary lymphoid organs

    rendering them undetectable in peripheral blood.

    These data suggest that irrespective of their phenotype CMV-specific T cells can confer

    protective immunity in situations of virus reactivation. Moreover, virus-reactivation appears

    54

  • to not only increase the size of the virus-specific T cell pool but also, in individuals that start

    off with "memory-type" CMV-specific T cells, induces a change from a predominant CD27+

    to a CD27' phenotype.

    a _ 0.0075-, a S-C 0.0050-> « S Ö O o S j l " 0.0025-O co w ü

    0.0000-

    0 100 200 300 400 500 600 0 1 2 3

    time (days after transplantation)

    Figure 2: Ongoing downmodulation of CD27 on CMV-specific CD8+ T cells after reactivation Figure 2a: Longitudinal analysis of CD27 expression on CMV-specific CD8+ T cells in relation to CMV-viral load in one representative patient with a CD28CD27TD45R0* phenotype of CMV-specific cells prior to CMV-reactivation. 2b: Percentage of CD27" CMV-tetramer' T cells in all patients during follow-up of reactivation. Time defined as t=0, before reactivation, t=l, during reactivation at peak PCR value, t=2, first timepoint after reactivation and t=3, > 6 months after reactivation

    The CD27" phenotype is correlated with the magnitude of the CD8+ T cell response

    during latency

    The above findings suggested a connection between the number of virus-specific T cells in

    latency and their phenotype which would accord with the expansion of CMV-specific

    CD8+CD27" T cells in immuno-compromised patients'0120, B-CLL patients21 and the elderly2" 23. Indeed, close analysis of the total percentage of CD8+ CMV-specific cells as determined

    by tetramer staining and the expression of CD27 by these cells shows a strong correlation

    between the loss of CD27 and a higher percentage of CMV-specific CD84 T cells as

    determined by tetramer staining, both for renal transplant recipients and healthy control

    individuals. The correlation of the percentage of CMV-tetramer+ CD8+ T cells and the

    percentage of CD27" of these cells is depicted in figure 3a, where it is shown that after the

    percentage of tetramer4" cells reaches approximately 1 %, > 50 % of these cells are CD27"

    whereas virtually all CMV-specific cells are CD27" when a percentage of 2 % or higher is

    reached (p=0.005). Also when depicted in absolute numbers, the amount of CMV-specific

    cells is linearly correlated to the amount of CD27" cells, figure 3b).

    55

  • a b

    - 5 r i —

    8 *s 5 005°

    £ > u

    5 10 15 20 % of CMV-tctramcrt cells

    0.00 0.01 0.02 0.03 0.04 0.05 0.06 0.07

    number of CMV-tetramer* cells (10''mL)

    Figure 3: High frequencies of CMV-specific CD27' effector ceils are correlated to the total amount of CMV-specific CD8* T cells. Figure 3a: Correlation of the percentage of total CMV-tetramer" T cells and the percentage CD27" of CMV-specific T cells. Closed circles: values in renal transplant recipients before or after reactivation; open circles: values in healthy control individuals, curve: functional relationship determined by:

    e**(0.226 + 0.86*ln(%tetramer» % CD27- = 100 * 1 + e**(0.226 + 0.86*ln(%tetramer)) p=0.005 Figure 3b: Correlation of the absolute number of total CMV-tetramer T cells and the absolute number CD27" CMV-specific T cells. Closed circles: values in renal transplant recipients, curve: linear relationship determined by: Absolute number of CMV-tetramer' cells = 0.001 + (1.46 * Absolute number CMV- tetramer'CD27") p=0.0012.

    Generation of the CD27" phenotype of virus-specific T cells is dependent on interaction

    of CD27 with its cellular ligand, CD70

    T cells downmodulate CD27 after interaction with its cellular ligand CD70 both in vitro and

    24; 25 24: 25 , In this process T cells receive co-stimulatory signals for expansion ' ~ and ,26 acquisition of effector functions" . To analyse if virus-induced CD70 expression could

    account for the down-modulation of CD27 on virus-specific T cells after CMV reactivation,

    CD70 expression was measured in the reactivating patients. Indeed, whereas during latency

    most CMV tetramer-binding T cells were CD70 negative (Figure 4 top panel left), CD70

    transiently increased during reactivation episodes (Figure 4 middle and right panel).

    Accordingly, viral antigen-induced CD70 expression could be induced in vitro and the

    magnitude of CD70 expression showed a dose-response relation with the virus-derived

    stimulus (Figure 4, bottom panel).To test if viral antigen-induced upregulation of CD70

    dictated CD27 expression, T cells from donors with a CD45R0CD27' phenotype (Figure

    5a) were cultured for five days with specific peptide and CMV-antigen to activate helper T

    cells in the absence and presence of blocking CD70 mAb. In these cultures, CMV-specific T

    cells rapidly downregulated CD27 (Figure 5b). However, addition of CD70 mAb to these

    56

  • " ifl

    t = 0

    2.1

    t = l t = 2

    Tetramer

    Figure 4: CD70 expression is induced by antigen

    and is dose dependent Figure 4a:CD70 is expressed on CD8 ' and CMV-specific CD8* T lymphocytes during viral replication in vivo. All plots gated on C D 8 ' T cells. Time defined as t=0, before reactivation, t= l , during reactivation t=2, after reactivation, quadrant percentages depicted as percentage CD70' of tetramcr* cells.Figure 4b: Left panel: Upon in vitro stimulation with CMV-antigen and CMV-peptide, CD70 expression is induced on CD8* T cells; total CD8+ T cells plotted in gray, CMV-specific CD8 ' T cells plotted in black. Right panel: Expression levelsof CD70 on CD8* T cells in various doses of CMV-antigen and peptide stimulation. X-axis: Log fluorescence CD70; Filled histogram: no stimulus, closed histogram: stimulation with 0.15625 uL/ mL CMV-antigen and 0.0195 ugr/mL CMV-peptidc, dotted histogram: stimulation with 0.625 uL/ mL CMV-antigen and 0.0781 ugr/mL CMV-peptide, bold histogram: stimulation with 2.5 uL/ mL CMV-antigen and 0.3125 ugr/mL CMV-peptide.

    CFSF. CD70

    Ji4r^ Mr'

    CD27

    Lymphocytes

    C CD70

    CD70 block Peptide +

    CMV-antigen

    Tetramer CD27

    Figure 5: CD70 induces differentiation of CD27* CMV-specific CDS* T cells Figure 5a: phenotype of CMV-specific CD8' T cells before stimulation, x-axis log fluorescence CD27, y-axis log fluorescence CD45RA. Figure 5b: Left panel: expression of CD70 on total lymphocytes after stimulation with CMV-peptide and CMV-antigen (open histogram) and no stimulus (filled histogram). X-axis: log fluorescence CD70. Right panel: Dot plots of CD27 expression on tetramer* CD8" T cells after 5 days of stimulation with peptide and CMV-antigen with or without blocking of CD70. Plots gated on CD8' T cells x-axis: log fluorescence CMV-tetramer y-axis: log fluorescence CD27; Histogram overlay of CD27 expression on CMV-specific CD8~ T cells without blocking of CD70 (filled histogram) and with blocking of CD70 (open histogram).

    cultures reversed this blocking effect and yielded CD27+ CMV-specific T cells at day 5 of

    culture. Helper CD4f T cells can directly support CD8+ T cell expansion through secretion of

    stimulatory cytokines and provision of membrane-bound helper factors"7. Furthermore, the

    homeostatic cytokine IL-15 was shown to be involved in maintenance and

    activation/functional differentiation of memory CD8T T cells28"30. To analyse a possible

    qualitative role of different cytokines on CD8+ T cell differentiation, the effects of cytokines,

    57

  • Lymphocytes Peptide

    CD70 block

    CD70 Tetramer CD27

    Figure 6: CD7Ö expression is induced by IL-2 and IL-15 but not by IL-21. Figure 6a-c: Left panels: expression of CD70 on total lymphocytes after stimulation with CMV-peptide and IL-2, IL-15 or 11-21 respectively (open histogram) and no stimulus (filled histogram). X-axis: log fluorescence CD70. Right panels: Dot plots of CD27 expression on tetramer" CD8~ T cells after 5 days of stimulation with peptide and IL-2, IL-15 or IL-21 respectively with or without blocking of CD70. Plots gated on CD8^ T cells x-axis: log fluorescence CMV-tctramer; y-axis: log fluorescence CD27: Histogram overlays of CD27 expression on CMV-specific CD8' T cells with different stimulations without blocking of CD70 (filled histogram) and with blocking of CD70 (open histogram).

    which previously were found to support proliferation of CMV-specific T cells16, on CD70 and

    CD27 expression were analysed. In conjunction with peptide, IL-2 and IL-15 induced CD70

    expression on the CMV-specific T cells and CD70-dependent down-modulation of CD27 (Fig

    6 a and b). Though IL-21 was reported to only stimulate CD45RA' thymocytes31, in our

    experiments, in an antigen-specific setting, also CD45R0+CD8+ T cells were responsive to IL-

    21. Interestingly, IL-21, a cytokine presumed to limit effector cell expansion32, did not induce

    CD70 expression and supported propagation of CD27+ CMV-specific T cells without

    differentiation Fig 6c).

    DISCUSSION

    Our results show that, in persistent viral infection, reactivation increases the number of

    memory cells and skews these cells to a CD27" phenotype. In primary infection, the amount

    58

  • of antigen determines the size of the murine CD8+ T cell response. Subsequently, the

    magnitude of this CD8+ T cell response sets the size of the antigen-specific T cell pool during

    the memory phase33'34. Recently it was demonstrated that the contraction phase of the primary

    immuneresponse is independent of the clearance of antigen35. However, both cytokines

    present during initial priming and the amount of antigen present after the contraction phase

    are likely to have an impact on the quality and quantity of the memory cells rendered

    thereafter34"36. It was proposed that different viruses elicit different human CD8+ T cell

    responses with respect to their surface phenotype9. EBV and HIV predominantly elicit CD28" +CD27+CD8+ responses. The analysis of antigen-specific memory CD8! T cells directed

    against influenza virus37; 38, which is completely cleared from the host, reveals that these cells

    are all CD28+CD27^ and present at low percentages compared to the frequency of CD8+ T

    cells directed at persistent viruses. CMV-specific responses in both healthy individuals and

    renal transplant recipients vary in magnitude and phenotype among individuals ' . The

    strong correlation between the percentage of virus-specific cells in the total CD8+ T cell

    population and the percentage of CD27* cells of the virus-specific cells indicates that a

    uniform mechanism takes place that drives both the expansion of virus-specific memory cells

    and their differentiation to cytotoxic "effector-type" cells.

    In this study, replication of CMV, measured by PCR, could be pinpointed to a very limited

    period shortly after transplantation, whereas the increase in total CMV-specific cells and the

    down-modulation of CD27 on these cells was an ongoing process for upto more than a year

    after transplantation. The ongoing rise in CMV-specific CD8+ T cells therefore is possibly

    generated by an increase in the number of infected host cells that persistently present antigen

    to immune cells. The only known mechanism through which CD27 is downregulated is after

    interaction with its ligand CD70. CD70 belongs to the TNF family and its expression on B

    and T cells can be induced by antigenic stimulation in vitro 39 and in vivo (this study) and

    appears to be modulated by the cytokine milieu. Indeed, our study showed that TCR-

    triggering in combination with IL-2 and IL-15 induced CD70, whereas IL-21 did not give

    CD70 expression. Accordingly, IL2 and IL-15 induced expansion of CD27" virus-specific T

    cells whereas IL-21 promoted outgrowth of CD27+ cells. Though both IL-2 and IL-21 are

    helper cell derived cytokines, the differentiation of CMV-specific memory CD8~ T cells to a

    CD27" phenotype upon activation with whole CMV-antigen in combination with peptide

    59

  • shows that the role of IL-21 production by CMV-specific CD4' memory T cells, reported to

    have a Thl profile , is limited in accordance with previous reports where IL-21 was shown to

    be a Th2 cytokine41. Whereas production of helper-cell derived cytokines as IL-21 and IL-2 is

    limited to the antigenic activation phase, IL-15 can be produced by a number of lymphoid and

    non-lymphoid cells42. It is well feasible that viruses elicit distinct helper cell cytokine profiles

    by differential infection of APC and by different target cell tropism, thereby also determining

    the number and phenotype of CD8' T cells during the memory phase43"45.

    CD27-deficient mice have a reduced ability to form adequate numbers of antigen-specific T

    cells upon viral infection46. In contrast, mice that constitutively express CD70 on B cells have

    a strongly enhanced capacity to form effector T cells in response to viruses, tumours and

    protein antigens47 (R. Arens, manuscript in preparation). Thus these mouse models strongly

    infer that the CD27-CD70 interaction is a major determinant in setting the size of the antigen-

    specific pool after immunization. The way in which CD27 signalling regulates these

    processes is incompletely unravelled but CD27"'" T cells show diminished survival after

    stimulation46. An effect of CD27 on T-cell survival would be in line with proposed functions

    of related TNF-R family molecules such as OX40 and 4-IBB that increase expression of anti-

    apoptotic molecules such as Bcl-2 and Bcl-xL48o°. Indeed, recently we found strong

    upregulation of Bcl-xL expression by CD27 triggering (M. van Oosterwij k. manuscript in

    preparation). On basis of these observations, it can be postulated that through pathogen-

    induced expression of CD70, and subsequent triggering of CD27, upregulation of Bcl-xL and

    increased survival of antigen-specific T cells, the contraction phase after initial expansion is

    altered leading to a higher setpoint in memory cell numbers. Simultaneously, through

    prolonged survival, T cells may receive differentiation signals for a longer period. This

    proposed link between survival and differentiation may explain the clear correlation we found

    between CMV-specific T cell numbers and their phenotype. In our in vitro studies we did not

    find an effect of CD70 blockade on numbers of antigen-specific T cells although these

    antibodies were capable to block CD27 signalling (figure 5 and 6). The lack of effect might

    be explained by the fact that upregulation of Bcl-xL may protect T cells from death by

    neglect, i.e. cytokine shortage, which may be essential in vivo but irrelevant in our cultures

    where ample cytokines are present.

    60

  • Taken together, our data imply that the induction and expansion of CD8 virus specific T cells

    and their differentiation to the CD27" effector phenotype is driven both by antigenic load and

    helper T cell-derived or homeostatic cytokines. We submit that regulation of CD70

    expression could be central in this process. Elucidating the differentials inducing and

    sustaining the CD27" effector phenotype could have profound impact on immunotherapeutic

    strategies, especially in disease situations where strong and persistent cytolytic responses are

    warranted.

    References 1. Sprent J, Surh CD: T cell memory. Annu.Rev.Immunol.2002;551-579. 2. Hamann D, Baars PA, Rep MH, et al. Phenotypic and functional separation of memory and effector human

    CD8+T cells. J.Exp.Med. 1997;186:1407-1418. 3. 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;401:708-712. 4. Ravkov EV, Myrick CM, Altman JD: Immediate Early Effector Functions of Virus-Specific

    CD8(+)CCR7(+) Memory Cells in Humans Defined by HLA and CC Chemokine Ligand 19 Tetramcrs. J Immunol.2003; 170:2461-2468.

    5. Ellefsen K, Harari A, Champagne P, Bart PA, Sekaly RP, Pantaleo G: Distribution and functional analysis of memory antiviral CD8 T cell responses in HIV-1 and cytomegalovirus infections. Eur.J lmmunol.2002;32:3756-3764.

    6. Weekes MP, Wills MR, Mynard K, Hicks R, Sissons JG, Carmichae! AJ: Large clonal expansions of human virus-specific memory cytotoxic T lymphocytes within the CD57+ CD28- CD8+ T-cell population. Immunology 1999;98:443-449.

    7. Hamann D, Kostense S, Wolthers KC. et al. Evidence that human CD8+CD45RA+CD27- cells are induced by antigen and evolve through extensive rounds of division. Int. Immunol. 1999; 11:1027-1033.

    8. Posnett DN, Sinha R, Kabak S, Russo C: Clonal populations of T cells in normal elderly humans: the T cell equivalent to "benign monoclonal gammapathy". J Exp.Med. 1994;179:609-618.

    9. Appay V, Dunbar PR. Callan M. et al. Memory CD8+ T cells vary in differentiation phenotype in different persistent virus infections. Nat.Med.2002;8:379-385.

    10. Gamadia LE, Rentenaar RJ, Baars PA, et al. Differentiation of cytomegalovirus-specific CD8(+) T cells in healthy and immunosuppressed virus carriers. Blood 2001;98:754-761.

    11. Roos MT, van Lier RA, Hamann D, et al. Changes in the composition of circulating CD8+ T cell subsets during acute epstetn-barr and human immunodeficiency vims infections in humans. J.Infect.Dis. 2000;182:451-458.

    12. Appay V, Papagno L, Spina CA, et al. Dynamics of T cell responses in HIV infection. J.Immunol.2002; 168:3660-3666.

    13. Gamadia LE, Remmerswaal EB. Weel JF. Bemelman F, van Lier RA, ten Berge IJ: Primary immune responses to human cytomegalovirus: a critical role for IFN{gamma (producing CD4+ T cells in protection against CMV-disease. Blood 2003;101:2686-2692.

    14. Pawelec G, Adibzadeh M, Rehbein A, Hahnel K, Wagner W, Engel A: In vitro senescence models for human T lymphocytes. Vaccine 2000; 18:1666-1674.

    15. Wills MR, Okecha G, Weekes MP. Gandhi MK, Sissons PJ, Carmichael AJ: Identification of naive or antigen-experienced human CD8(+) T cells by expression of costimulation and chemokine receptors: analysis of the human cytomegalovirus-specific CD8(+) T cell response. J Immunol.2002; 168:5455-5464.

    16. van Leeuwen EM, Gamadia LE, Baars PA, Remmerswaal EB, ten Berge IJ, van Lier RA: Proliferation requirements of cytomegalovirus-specific, effector-type human CD8+ T cells. J Immunol.2002; 169:5838-5843.

    61

  • 17. Champagne P. Ogg GS, King AS, et al. Skewed maturation of memory HIV-specific CD8 T lymphocytes. Nature 2001;410:106-111.

    18. Altman JD. Moss PA, Goulder PJ. et al. Phenotypic analysis of antigen-specific T lymphocytes. Science 1996;274:94-96.

    19. Boom R. Sol C, Weel JF. Gerrits Y, de Boer M. Wertheim-van Dillen PME: A highly sensitive assay for detection and quantitation of human cytomegalovirus DNA in serum and plasma by PCR and electrochemiluminescence. J Clin.Microbiol. 1999:37:1489-1497.

    20. Papagno L, Appay V. Sutton J, et al. Comparison between HIV- and CMV-specific T cell responses in long-term HIV infected donors. Clin.Exp.Immunol.2002;130:509-517.

    21. Mackus. WJ., Frakking, FN., Grummels A. et al. Expansion of CMV-specific CD8*CD45RA CD27" T cells in B-cell chronic lymphocytic leukemia. Blood 2003: advanced online publication

    22. Khan N. Shariff N. Cobbold M, et al. Cytomegalovirus seropositivity drives the CD8 T cell repertoire toward greater clonality in healthy elderly individuals. J Immunol.2002;169:1984-1992.

    23. Looney RJ. Falsey A, Campbell D, et al. Role of cytomegalovirus in the T cell changes seen in elderly-individuals. Clin.Immunol. 1999;90:213-219.

    24. Hintzen RQ, Lens SM, Lammers K. Kuiper H, Beckmann MP. Van Lier RA: Engagement of CD27 with its ligand CD70 provides a second signal forT cell activation. J.Immunol. 1995;154:2612-2623.

    25. Arens R, Tesselaar K, Baars PA. Van Schijndel GM, Hendriks J, Pals ST, Krimpenfort P. Borst J. van Ocrs MJ, Van Lier RA: Constitutive CD27CD70 interaction induces expansion of effector-type T cells and results in IFNy-mediated B cell depletion. Immunity 2001:15:801-812.

    26. Goodwin RG. Alderson MR. Smith CA. ct al. Molecular and biological characterization of a ligand for CD27 defines a new family of cytokines with homology to tumor necrosis factor. Cell 1993;73:447-456.

    27. Bourgeois C. Rocha B. Tanchot C: A role for CD40 expression on CD8+ T cells in the generation of CD8+ T cell memory. Science 2002;297:2060-2063.

    28. Mueller YM, Bojczuk PM. Halstead ES, et al. IL-15 enhances survival and function of HIV-specific CD8-1-T cells. Blood 2003;101:1024-1029.

    29. Becker TC, Wherry EJ. Boone D. et al. Interleukin 15 is required for proliferative renewal of virus-specific memory CD8 T cells. J Exp.Med.2002;195:1541-1548.

    30. Wherry EJ. Becker TC. Boone D, Kaja MK, Ma A, Ahmed R: Homeostatic proliferation but not the generation of virus specific memory CD8 T cells is impaired in the absence of IL-15 or IL-15Ralpha. Adv.Exp.Med.Biol.2002;512:165-175.

    31. Parrish-Novak J, Dillon SR, Nelson A. et al. Interleukin 21 and its receptor are involved in NK cell expansion and regulation of lymphocyte function. Nature 2000; 408:57-63.

    32. Kasaian MT. Whittcrs MJ. Carter LL, et al. IL-21 limits NK cell responses and promotes antigen-specific T cell activation: a mediator of the transition from innate to adaptive immunity. Immunity 2000; 16:559-569.

    33. Wherry EJ. McElhaugh MJ. Eisenlohr LC: Generation of CD8(-) T cell memory in response to low. high, and excessive levels of epitope. J.Immunol.2002; 168:4455-4461.

    34. Murali-Krishna K, Altman JD. Suresh M. et al. Counting antigen-specific CD8 T cells: a reevaluation of bystander activation during viral infection. Immunity. 1998;8:177-187.

    35. Badovinac VP, Porter BB, Harty JT: Programmed contraction of CD8(+) T cells after infection. Nat.Immunol.2002;3:619-626.

    36. Oh S. Berzofsky JA. Burke DS. Waldmann TA. Perera LP: Coadministration of HIV vaccine vectors with vaccinia viruses expressing IL-15 but not IL-2 induces long-lasting cellular immunity. Proc.Natl.Acad.Sci.U.S.A.2003:100:3392-3397.

    37. Pittet MJ, Valmori D, Dunbar PR. el al. High frequencies of naive Melan-A/MART-1-specific CD8(+) T cells in a large proportion of human histocompatibility leukocyte antigen (HLA)-A2 individuals. J.Exp.Med. 1999;190:705-715.

    38. He XS, Mahmood K. Maecker HT. ct al. Analysis of the Frequencies and of the Memory T Cell Phenotypes of Human CD8+ T Cells Specific for Influenza A Viruses. J Infect.Dis.2003; 187:1075-1084.

    39. Lens SM. Baars PA, Hooibrink B, van Oers MH. Van Lier RA: Antigen-presenting cell-derived signals determine expression levels of CD70 on primed T cells. Immunology 1997;90:38-45.

    40. Rentenaar RJ, Gamadia LE. van der Hoek N. et al. Development of virus-specific CD4(+) T cells during primary cytomegalovirus infection. J Clin.Invcst. 2000:105:541-548.

    62

  • 41. Wurster AL, Rodgers VL, Satoskar AR, et al. Interleukin 21 is a T helper (Th) cell 2 cytokine that specifically inhibits the differentiation of naive Th cells into interferon gamma-producing Thl cells. J Exp.Med.2002; 196:969-977.

    42. Grabstein KH, Eisenman J, Shanebeck K, et al. Cloning of a T cell growth factor that interacts with the beta chain of the interleukin-2 receptor. Science 1994;264:965-968.

    43. Ozaki K, Spolski R, Feng CG, et al. A critical role for IL-21 in regulating immunoglobulin production. Science 2002;298:1630-1634.

    44. de Boer OJ, Hirsch F, van der Wal AC, van der Loos CM, Das PK, Becker AE: Costimulatory molecules in human atherosclerotic plaques: an indication of antigen specific T lymphocyte activation. Atherosclerosis 1997;133:227-234.

    45. Bennett F, Luxenberg D, Ling V, et al. Program death-1 engagement upon TCR activation has distinct effects on costimulation and cytokine-driven proliferation: attenuation of ICOS, IL-4, and IL-21, but not CD28, IL-7, and IL-15 responses. J Immunol.2003; 170:711-718.

    46. Hendriks J. Gravestein LA, Tessclaar K, Van Lier RA, Schumacher TN, Borst J: CD27 is required for generation and long-term maintenance of T cell immunity. Nat.Immunol.2000; 1:433-440.

    47. Tesselaar K, Arens R, van Schijndel GM, et al. Lethal T cell immunodeficiency induced by chronic costimulation via CD27-CD70 interactions. Nat.Immunol.2003;4:49-54.

    48. Rogers PR, Song J, Gramaglia I, Killeen N, Croft M: OX40 promotes Bcl-xL and Bcl-2 expression and is essential for long-term survival of CD4 T cells. Immunity 2001;15:445-455.

    49. Bertram EM. Lau P, Watts TH: Temporal segregation of 4-1BB versus CD28-mediated costimulation: 4-1BB ligand influences T cell numbers late in the primary response and regulates the size of the T cell memory response following influenza infection. J Immunol.2002; 168:3777-3785.

    50. Lee HW, Park SJ, Choi BK, Kim HH, Nam KO, Kwon BS: 4-IBB promotes the survival of CD8+ T lymphocytes by increasing expression of Bcl-xL and Bfl-1. J Immunol.2002:169:4882-4888.

    63