After acute infection Epstein-Barr virus (EBV)–specific memory CD8+ T cells exit cell cycle, and a proportion of these antigen-experienced cells reexpress CD45RA (CD45 which predominantly express exon A). However, the signals involved are not known. We investigated the roles of interleukin 15 (IL-15) and interferon-α/β (IFN-I) in these processes, since these mediators have a crucial but undefined role in the maintenance of CD8+ T-cell memory. We show that IFN-I (but not IL-15) allows activated EBV-specific CD8+ T cells to leave cell cycle without entering apoptosis. This was associated with up-regulation of the cyclin inhibitor p27, but not of CD45RA. In contrast, IL-15 (but not IFN-I) induced “homeostatic” proliferation and CD45RA reexpression by these cells in vitro. Different signals, therefore, induce quiescence and CD45RA reexpression in activated EBV-specific CD8+ T cells. After T-cell receptor (TCR) activation freshly isolated CD45RA+ antigen-experienced CD8+ T cells show poor proliferative activity but are highly cytotoxic and secrete IFN-γ efficiently. This suggests functional reprogramming toward effector function but away from proliferation. The induction of quiescence and the generation of proliferation-independent effector CD8+ T cells that reexpress CD45RA may minimize the impact of replicative senescence in virus-specific populations that would otherwise occur during decades of persistent infection.

An immune response to antigen induces the activation and expansion of specific T lymphocytes, followed by the clearance of the majority of these cells when the antigen is eliminated.1  However, the rescue of some antigen-specific T cells from apoptosis is essential for the maintenance of a memory T-cell pool.2  Herpes viruses, such as Epstein-Barr virus (EBV)3  and cytomegalovirus (CMV),4  induce persistent latent infection. The memory T-cell pool for such viruses will be subjected to repeated stimulation in response to viral reactivation in vivo. Since the repeated stimulation of T lymphocytes leads to progressive telomere erosion that ultimately leads to growth arrest or replicative senescence,5,6  factors capable of actively inducing quiescence in memory T cells between episodes of virus reactivation are crucial for long-term maintenance of T-cell memory.3,7 

A feature of memory T cells that are specific for persistent viruses such as EBV and CMV is the emergence of an antigen-experienced CD8+ T-cell population that reexpresses CD45RA (CD45 which predominantly express exon A).8  These CD45RA reexpressing T cells also accumulate in elderly individuals.9  Some studies have suggested that these cells are close to end-stage differentiation.10,11  However, more recent reports indicate that they can be reactivated to proliferate,7,8,12  although to a lesser extent than other T-cell subsets13  and represent a stable memory population.8,14  As CD8+ T cells that reexpress CD45RA accumulate during chronic viral infections, they are likely to be involved in the long-term preservation of immunity to agents that induce life-long infection.3,4  However, the nature of factors that induce quiescence and/or CD45RA reexpression in human virus-specific CD8+ T cells and the functional role of these cells have not been defined.

The relative expression of positive and negative regulators of cell cycle in human memory T cells is poorly characterized. Cyclins D (D1, D2-D3), represent positive regulators of proliferation, but only cyclins D2 and D3 are found in human T cells.15  Cyclins are required to form complexes with cyclin-dependent kinases (cdk's) to function. During G1 phase progression, these cyclin/cdk complexes phosphorylate members of the pocket protein family, leading to the release of the transcription factor E2F.16  This results in the transcription of genes required for progression from G1 to S-phase of the cell cycle.17  Cell-cycle progression is repressed by cdk inhibitors (CKIs) such as p27, which is a member of the cycle inhibitor 21 and kinase inhibitor p27 (Cip/Kip) family of inhibitors.18  In this study we sought to determine whether the induction of quiescence in activated virus-specific CD8+ T cells is due to down-regulation of the cyclins or up-regulation of cyclin inhibitors or both.

To identify factors that induce either quiescence or CD45RA reexpression in human CD8+ T cells, we activated and expanded EBV-specific CD8+ CD45RO+ T cells with viral peptides in vitro as a model of chronic viral infection in humans. Interferon α (IFN-α) and IFN-β belong to the type I IFN family, signal via the same receptor, and, thus, have been shown to share the same antiproliferative, antiapoptotic, and antiviral characteristics.19-22  We will refer to these cytokines collectively as IFN-I throughout this manuscript. We investigated the ability of IFN-I and also interleukin 15 (IL-15) to promote survival, quiescence, and/or CD45RA reexpression in these cells. We show that, while IFN-I induces quiescence, only IL-15 induces CD45RA reexpression. Furthermore, CD45RA reexpressing EBV-specific CD8+ T cells represent a population that has been functionally refocused to mediate effector function without excessive proliferation. This would be crucial for the maintenance of specific cells during life-long stimulation by chronic infective agents.

Sample collection and preparation

Heparinized peripheral blood was collected from patients with acute infectious mononucleosis (AIM), from patients who recovered from AIM at least 10 years previously (chronic phase), and from healthy donors with no history of AIM. Peripheral blood mononuclear cells (PBMCs) were isolated by Ficoll density centrifugation and cryopreserved in liquid nitrogen. Purified CD8+ T-cell subsets were separated from PBMCs by negative selection using the VARIO MACS (magnetic-activated cell sorting) system (Miltenyi Biotec, Surrey, United Kingdom) as described in detail elsewhere.23  Approval was obtained from the Royal Free and University College Medical Ethics Group, University College London institutional review board for these studies. Informed consent was provided according to the Declaration of Helsinki.

Peptide-human leukocyte antigen (HLA) class I tetramers

Soluble phycoerythrin (PE)–labeled peptide-HLA class I tetramers (Proimmune, Oxford, United Kingdom) containing the HLA-B8–restricted peptide ligand RAKFKQLL from the EBV protein BZLF1 (abbreviated to RAK) were used to identify EBV lytic epitope-specific CD8+ T cells.24 

Stimulation and maintenance of lytic epitope-specific CD8+ T-cell lines

Specific CD8+ T-cell lines were generated by adding RAK peptide (Alta Bioscience, The University of Birmingham, United Kingdom) to freshly isolated PBMCs of individuals with chronic infection and who were HLA-B8 positive. Cells were incubated for 3 days at 37°C at which point 20 U/mL exogenous human recombinant IL-2 (R&D Systems, Abingdon, United Kingdom) was added. Cells received fresh complete medium (RPMI, 100 IU penicillin, 100 μg/mL streptomycin, 2 mM l-glutamine, and 10% fetal calf serum [Life Technologies, Paisley, United Kingdom]) and 20 U/mL exogenous IL-2 every 3 days. RAK-specific CD8+ T cells were restimulated every 14 days.

Flow cytometric analysis

Freshly isolated PBMC samples were analyzed by flow cytometry using PE-conjugated HLA class I-peptide tetrameric complexes (Pro-immune) as previously described.8  Specific antibodies used included anti-CD3 and anti-CD8 (Dako, Cambridgeshire, United Kingdom), anti-CD45RA and anti-CD45RO (BD Biosciences, Oxford, United Kingdom). Staining of intracellular proteins has been described elsewhere.8  Immunoglobulin G1 (IgG1) isotype controls were used in all experiments.

Cell-cycle analysis

Cells were stained with anti-CD8 and RAK tetramer (Pro-immune), permeabilized with Fix and Perm reagent (CALTAG-Medsystems, Silverstone, United Kingdom), and then stained with anti-Ki67 FITC (Dako). Appropriate IgG1 isotype antibodies as well as resting PBMCs were used as negative controls.

Cell-cycle analysis was also determined by 7-aminoactinomycin D (7AAD) staining of DNA; freshly isolated PBMCs were labeled with RAK tetramer and other antibodies to identify particular T-cell subsets. Cell suspensions were permeabilized using Cytofix/Cytoperm (BD Biosciences). After incubation at 4°C for 15 minutes in Cytofix/Cytoperm and 2 further washes, 7AAD (40 μg/mL) was added to cell pellets. Noncycling, resting PBMCs were used as negative controls.

Loss of carboxy-fluorescein diacetate, succinimidyl ester (CFSE; Sigma-Aldrich, Gillingham, United Kingdom) staining was also used as a marker of proliferation. T cells were washed twice and resuspended in 1 mL phosphate-buffered saline. CFSE (0.5 μM/mL) was added, and cells were incubated for 5 minutes at room temperature then washed in PBS.

Rescue from apoptosis

The rescue of T cells from cytokine-dependent death has been described in detail previously.8  Fibroblast-conditioned medium, a physiologic source of IFN-β, was used to prevent apoptosis.19,20  Viable cell recovery (VCR) was determined by trypan blue dye exclusion after 24 hours.

CD45RO to CD45RA reversion

EBV-specific CD8+ T cells were activated with peptide-pulsed autologous antigen-presenting cells (APCs), and maintained in culture for 3 weeks. T cells were washed and assessed for CD45 expression by flow cytometric analysis. T cells (2 × 106) were subsequently added to 24-well inserts with a 0.4-μm pore diameter (BD Biosciences; Discovery Labware, Oxford, United Kingdom), which were then placed into wells of a 24-well plate containing confluent human embryonic lung fibroblasts as a source of IFN-β.19,20  The cultures were replenished with complete medium every 2 days. CD45 expression and VCR were assessed up to 19 days later.

CD8+ CD45RO+ T cells were purified as already described and cultured with 20 U/mL exogenous IL-15 (R&D Systems). CD45RA reexpression and cell-cycle progression were determined by flow cytometry 7 and 13 days later.

Functional assessment of EBV-specific CD8+ T cells and CD8+ T-cell subsets

EBV-specific CD8+ T cells that were cultured with either medium, IL-2, IFN-2α, or IFN-β were restimulated as described in maintenance of T-cell lines. After 2 hours, 5 μg/mL Brefeldin A (Sigma-Aldrich) was added, and cells were incubated overnight at 37°C. These cells were subsequently washed and stained for surface antigens and then for intracellular IFN-γ as described.25  Perforin expression in these cells was also determined 5 days after restimulation. Five days after reactivation and after staining with surface antigen, T cells were permeabilized, stained with anti-Perforin–fluorescein isothiocyanate (FITC; BD Biosciences), and fixed.

CD8+ T-cell subsets were sorted using a MoFlo cell sorter on the basis of CD27 and CD45RA expression. All CD8+ T-cell subsets were activated with a concentration range of anti-CD3 and irradiated autologous APCs. Proliferation was determined by tritiated thymidine incorporation 72 hours later. To determine the cytotoxic potential of CD8+ T cells, sorted T cells were stimulated with a concentration range of anti-CD3 plus exogenous APCs in conjunction with antibodies directed against CD107 A and B.26  Cells were incubated for 1 hour at 37°C after which Monensin A 100 μM (Sigma-Aldrich) was added, and cells were incubated for a further 4 hours at 37°C. Cells were stained for other surface markers at this point.

Western blot analysis

Whole-cell extracts were prepared by lysing cells with 4 times the packed cell volume of lysis buffer (1% Nonidet P-40 ([Octylphenoxy]polyethoxyethanol), 100 mM NaCl, 20 mM Tris (tris(hydroxymethyl)aminomethane)–HCl pH 7.4, 10 mM NaF, 1 mM Na3VO4, and protease inhibitors; Roche, Lewes, United Kingdom) on ice for 15 minutes. Protein yield was quantified by Bio-Rad Dc protein assay kit (Bio-Rad, Hemel Hempstead, United Kingdom). Lysate (50 μg) was separated by sodium dodecyl sulfate (SDS)–polyacrylamide gel electrophoresis and transferred to nitrocellulose membranes (Technopath, Limerick, Ireland), and specific proteins were detected using antibodies. The monoclonal antibodies against cyclin D2 and cyclin D3 antibody were acquired from NeoMarkers (Union City, CA). The actin, glyceraldehyde-3-phosphate dehydrogenase (GAPDH), and p27 antibodies were from Santa Cruz Biotechnology (Santa Cruz, CA). The antibodies were detected using horseradish peroxidase–linked goat anti–mouse or anti–rabbit IgG (Dako) and visualized by the enhanced chemiluminescent (ECL) detection system (Amersham Biosciences, Chalfont St. Giles, United Kingdom). Normalization ratios were calculated by dividing the optical density value of the antibody-positive protein band by that of the associated β-actin control band. Optical density values were calculated using Quantity One software (Bio-Rad)

Figure 1.

In vitro generation of EBV-specific CD8+ T cells. Peripheral blood mononuclear cells were isolated from donors with AIM (A) and from those who had recovered from AIM at least 1 year previously (B). The percentage of CD8+ EBV peptide (RAK)–specific T cells was determined by using HLA class I tetramers. (C) The cell-cycle status of antigen-specific T cells isolated during the acute phase of infection (open histogram) and those cells isolated from donors with chronic infection (filled histogram) were determined by staining cells with antibodies directed against Ki67. PBMCs isolated from donors with chronic infection (D) were stimulated in vitro with RAK peptide-pulsed autologous APCs plus exogenous IL-2 and maintained in culture for 4 weeks (E). (F) Proliferation was assessed by Ki67 staining in freshly isolated CD8+ tetramer+ cells (filled histogram) and in specifically activated CD8+ tetramer+ cells that were expanded for 1 month in vitro (open histogram). These results are representative of 6 separate experiments. (A, B, D, E) Numbers refer to percentages of tetramer-positive CD8+ cells.

Figure 1.

In vitro generation of EBV-specific CD8+ T cells. Peripheral blood mononuclear cells were isolated from donors with AIM (A) and from those who had recovered from AIM at least 1 year previously (B). The percentage of CD8+ EBV peptide (RAK)–specific T cells was determined by using HLA class I tetramers. (C) The cell-cycle status of antigen-specific T cells isolated during the acute phase of infection (open histogram) and those cells isolated from donors with chronic infection (filled histogram) were determined by staining cells with antibodies directed against Ki67. PBMCs isolated from donors with chronic infection (D) were stimulated in vitro with RAK peptide-pulsed autologous APCs plus exogenous IL-2 and maintained in culture for 4 weeks (E). (F) Proliferation was assessed by Ki67 staining in freshly isolated CD8+ tetramer+ cells (filled histogram) and in specifically activated CD8+ tetramer+ cells that were expanded for 1 month in vitro (open histogram). These results are representative of 6 separate experiments. (A, B, D, E) Numbers refer to percentages of tetramer-positive CD8+ cells.

Close modal

Statistics

The Student t test was used to determine the significance of the results.

Figure 2.

IFN-I can actively promote quiescence in virus-specific CD8+ T cells. Cell-cycle progression in activated CD8+ EBV tetramer+ T cells and in noncycling resting PBMC control cells was determined by tritiated thymidine incorporation (A). Active EBV virus-specific CD8+ EBV-tetramer+ T cells were subsequently cultured in fresh medium alone or fresh medium supplemented with exogenous IL-15 or IFN-I. Proliferation and cell survival were determined 4 days later by tritiated thymidine incorporation (B) and trypan blue exclusion (C), respectively (dotted line represents 100% survival). These experiments are representative of 5 separate experiments. cpm indicates counts per minute. Error bars represent standard error from the mean.

Figure 2.

IFN-I can actively promote quiescence in virus-specific CD8+ T cells. Cell-cycle progression in activated CD8+ EBV tetramer+ T cells and in noncycling resting PBMC control cells was determined by tritiated thymidine incorporation (A). Active EBV virus-specific CD8+ EBV-tetramer+ T cells were subsequently cultured in fresh medium alone or fresh medium supplemented with exogenous IL-15 or IFN-I. Proliferation and cell survival were determined 4 days later by tritiated thymidine incorporation (B) and trypan blue exclusion (C), respectively (dotted line represents 100% survival). These experiments are representative of 5 separate experiments. cpm indicates counts per minute. Error bars represent standard error from the mean.

Close modal

Expansion of EBV-specific T cells in vitro

There is considerable expansion of EBV lytic epitope-specific CD8+ T cells during infectious mononucleosis (AIM; Figure 1A). However, once the infection resolves, the virus-specific T-cell population contracts (Figure 1B). While the majority of EBV-specific CD8+ T cells are in cycle during AIM, the same EBV-specific memory populations are largely quiescent (Figure 1C). To investigate factors that could induce the quiescence in these populations, we stimulated EBV-specific CD8+ T cells from individuals with chronic infection (Figure 1C) with specific peptide (RAK)–pulsed autologous APCs (Figure 1D). After 2 stimulations over 4 weeks of culture, the percentage of CD8+ T cells that were specific for RAK increased from 0.6% (Figure 1D) to 96% (Figure 1E). In addition, before stimulation in vitro less than 3% of these cells were Ki67+, while greater than 80% of cells that were maintained in culture for 4 weeks expressed this marker (Figure 1F).

IFN-I can inhibit cell-cycle progression in activated EBV-specific CD8+ T cells

We confirmed that the in vitro–expanded EBV-specific cells were in cycle by tritiated thymidine incorporation (Figure 2A). Peptide was removed by washing the cells, and the ability of either IL-15 or IFN-I to induce quiescence and or survival after 4 days in culture was determined. The virus-specific CD8+ T cells that were cultured in IL-15 were highly proliferative (Figure 2B) and showed good viable cell recovery, due in part to the cell expansion (Figure 2C). In contrast, virus-specific CD8+ T cells that were incubated in medium alone or those that were cultured in IFN-I were not actively cycling (Figure 2B). However, there was poor survival in cells that were cultured in medium alone compared with those cultured in the presence of IFN-I (Figure 2C). These results show that, while survival of virus-specific CD8+ T cells can be promoted by both IL-15 and IFN-I, only the latter cytokines were capable of inducing quiescence. These findings are in agreement with our previous studies in which both IL-15 and IFN-I were shown to inhibit apoptosis in EBV-specific CD8+ T cells isolated from patients with AIM.27,28  However, in accordance with the results described in this report, previous reports have shown that only IFN-I was capable of inducing quiescence in activated T cells.27,28 

Virus-specific CD8+ T cells rescued by IFN-I or IL-15 can be reactivated functionally by antigen

It was not clear whether virus-specific CD8+ T cells that were maintained in IL-15 or IFN-I retain their ability to be reactivated by antigen. We, therefore, restimulated EBV-specific CD8+ T cells that had been incubated in medium alone or fresh medium supplemented with IL-15 or IFN-I for 4 days with RAK-pulsed irradiated APCs plus exogenous IL-2. We found that cells that had been cultured in all the different conditions could be induced to proliferate (Figure 3A) and up-regulated both IFN-γ (Figure 3B) and perforin (Figure 3C). These results indicate that irrespective of whether virus-specific CD8+ T cells are maintained in a resting or an activated state, they retain their capacity to react functionally to antigenic rechallenge.

Figure 3.

Reactivation of virus-specific CD8+ T cells rendered quiescent by IFN-I. EBV-specific T cells maintained in fresh medium alone, IL-15, IFN-α, or IFN-β were reactivated with peptide-pulsed autologous APCs plus exogenous IL-2. Proliferation was determined by tritiated thymidine incorporation 5 days later (A). Error bars refer to the standard error from the mean. IFN-γ (B) and perforin (C) expressions were determined 18 hours and 3 days after reactivation, respectively (gray histograms represent unstimulated control). These results are representative of 3 separate experiments. (B-C) Percentages shown refer to the proportion of cytokine-secreting T cells.

Figure 3.

Reactivation of virus-specific CD8+ T cells rendered quiescent by IFN-I. EBV-specific T cells maintained in fresh medium alone, IL-15, IFN-α, or IFN-β were reactivated with peptide-pulsed autologous APCs plus exogenous IL-2. Proliferation was determined by tritiated thymidine incorporation 5 days later (A). Error bars refer to the standard error from the mean. IFN-γ (B) and perforin (C) expressions were determined 18 hours and 3 days after reactivation, respectively (gray histograms represent unstimulated control). These results are representative of 3 separate experiments. (B-C) Percentages shown refer to the proportion of cytokine-secreting T cells.

Close modal

Cell-cycle associated protein signatures in resting and activated CD8+ T cells

It was not clear whether the induction of quiescence in activated virus-specific CD8+ T cells was a result of changes in the expression levels of cyclins or CKIs. To address this, we first investigated the expression of 2 key cyclins, namely cyclin D2 and D3, and also the CKI p27 in freshly isolated resting CD8+ T cells and activated CD8+ T cells from a patient with AIM that had been activated in vivo8  (Figure 4A). Freshly isolated CD8+CD45RA+ and CD8+CD45RO+ T cells expressed high levels of p27 (Figure 4A). While CD8+CD45RA+ T cells did not express cyclins D2 or D3, CD8+CD45RO+ T cells showed low cyclin D2 expression. In contrast, CD8+ T-cell populations from patients with AIM showed high levels of cyclin D3, low levels of cyclin D2, and considerably decreased expression of p27 compared with resting populations. The cyclin signature of activated EBV-specific CD8+ T cells that were expanded by specific peptide in vitro (Figure 4A, lane 3) resembled that of the same populations activated in patients with AIM in vivo (Figure 4B, lane 1). There was up-regulation of cyclin D3, low to negative cyclin D2, and considerably down-regulated p27 in these cells.

Figure 4.

Cell-cycle–associated protein signatures. (A) Proteins (cyclin D2/D3, p27, and actin) extracted from freshly isolated naive T cells from a noninfected donor (lane 1), freshly isolated memory T cells from a healthy noninfected individual (CD8+ CD45RO+) (lane 2), and PBMCs isolated from a donor with AIM (lane 3) were subjected to Western blot analysis. (B) Western blot analysis and normalization ratio calculating the β-actin/antibody-positive protein band of activated EBV-specific T cells (lane 1) which were removed from TCR stimulus and maintained in either fresh medium alone (lane 2), IL-15 (lane 3), or IFN-α (lane 4) for 4 days. These results are representative of 3 separate experiments.

Figure 4.

Cell-cycle–associated protein signatures. (A) Proteins (cyclin D2/D3, p27, and actin) extracted from freshly isolated naive T cells from a noninfected donor (lane 1), freshly isolated memory T cells from a healthy noninfected individual (CD8+ CD45RO+) (lane 2), and PBMCs isolated from a donor with AIM (lane 3) were subjected to Western blot analysis. (B) Western blot analysis and normalization ratio calculating the β-actin/antibody-positive protein band of activated EBV-specific T cells (lane 1) which were removed from TCR stimulus and maintained in either fresh medium alone (lane 2), IL-15 (lane 3), or IFN-α (lane 4) for 4 days. These results are representative of 3 separate experiments.

Close modal

To simulate the effects of immune resolution, we removed IL-2 and peptide from the EBV-specific CD8+ T cells that were generated in vitro and examined the expression of cell-cycle regulators (Figure 4B, lane2). After 4 days of culture, cells were not in cycle and showed down-regulation of cyclin D3 and up-regulation of p27. However, in the absence of exogenous cytokines there was considerable apoptosis that eventually led to the loss of all cells (see Figure 2). This indicates that additional signals are required for the persistence of these cells. As shown in Figure 2, both IL-15 and IFN-I promote survival of EBV-specific CD8+ T cells; however, the cyclin profiles of cells rescued by either group of mediators were quite distinct. Maintenance in IFNα–induced quiescence associated with down-regulation of cyclin D3 and up-regulation of p27 (Figure 4B, lane 4), while maintenance in IL-15 induced cell cycle associated with high cyclin D3 levels and low p27 expression (Figure 4B, lane 4). Normalization ratios (based on optical densities) of antibody-positive protein bands versus β-actin controls were calculated to normalize the differences in β-actin expression between samples examined (Tables 1 and 2) support the results described here. It is possible that up-regulation of p27 is due only to growth factor removal, since elevated p27 levels were also found in cells maintained in fresh medium alone (Figure 4B, lane 2). However, previous studies have shown that IFN-α can also directly induce p27.21,29 

Table 1.

Normalization ratios (based on optical densities) of antibody-positive protein bands versus β-actin controls




Naive T cells

Memory T cells

AIM
Cyclin D2  0.32   0.76   0.40  
Cyclin D3  0.25   0.39   0.95  
p27
 
1.55
 
0.89
 
0.81
 



Naive T cells

Memory T cells

AIM
Cyclin D2  0.32   0.76   0.40  
Cyclin D3  0.25   0.39   0.95  
p27
 
1.55
 
0.89
 
0.81
 
Table 2.

Normalization ratios (based on optical densities) of antibody-positive protein bands versus β-actin controls




Activated EBV-specific T cells

Medium control

EBV-specific T cells and IL-15

EBV-specific T cells and IFN-α
Cyclin D2  0.09   0.14   0.13   0.11  
Cyclin D3  0.52   0.30   0.43   0.20  
p27
 
0.40
 
1.17
 
0.57
 
0.96
 



Activated EBV-specific T cells

Medium control

EBV-specific T cells and IL-15

EBV-specific T cells and IFN-α
Cyclin D2  0.09   0.14   0.13   0.11  
Cyclin D3  0.52   0.30   0.43   0.20  
p27
 
0.40
 
1.17
 
0.57
 
0.96
 

IFN-I does not induce CD45RO to CD45RA reversion by EBV-specific CD8+ T cells

Since virus-specific memory CD8+ T cells that reexpress CD45RA are quiescent,8  we investigated whether induction of cell-cycle exit in these cells by IFN-I was linked to CD45RA reexpression (Figure 5). We removed both peptide and IL-2 from EBV-specific CD8+ CD45RO+ cells that were expanded in vitro and cocultured these cells in a dual chamber system with fibroblasts. We have performed extensive studies to show that fibroblast-derived IFN-I promotes activated T-cell survival without proliferation in a manner identical to that of exogenous IFN-I.20,30  Furthermore, blocking anti–IFN-I receptor (IFNR) antibodies inhibit the survival and quiescence-promoting effects of fibroblast-conditioned medium.20,25  The advantage of using fibroblasts in this coculture system is that we are investigating a physiologic source of this cytokine that is continuously secreted during the culture period and, therefore, does not need to be replenished during the experiment.

CD45RO+ expression, as expected, was found to be enhanced in both total CD8+ T cells (Figure 5A) and in EBV tetramer+ T cells (Figure 5B) isolated from the same patient with AIM, when compared with CD45RO+ expression in freshly isolated EBV-specific CD8+ T cells isolated from a patient with chronic infection (Figure 5C). The proportion of freshly isolated tetramer+ cells, isolated from a donor with chronic EBV infection, expressing CD45RO+, both CD45RO+ and CD45RA+, and CD45RA+ alone was 34%, 20%, and 46%, respectively (Figure 5C). After 28 days of culture with peptide and IL-2, 98% of these virus-specific T cells were CD45RO+ (Figure 5D), and 35% of these cells were in active cell cycle (Figure 5E). When these cells were washed and cocultured with fibroblasts for 7 days, 99% of cells retained CD45RO expression (Figure 5F), but only 0.4% (Figure 5G) was in the active phases of cell cycle. This showed that the induction of quiescence could be dissociated from CD45RA reexpression in virus-specific CD8+ T cells.

IL-15 but not IFN-I can induce CD45RO to CD45RA reversion in resting virus-specific CD8+ T cells

It has been shown previously that IL-15 can induce CD45RA reexpression in polyclonal populations of central memory CD8+ T cells (CD45RO+ CCR7+) but not from the more differentiated effector memory (CD45RO+, CCR7) populations.13  The EBV-specific populations that we expanded in vitro and used in the previous experiments were highly differentiated and of the effector memory type (not shown). It was, therefore, possible that the lack of CD45RA reexpression observed in the previous experiments was due to extensive differentiation beyond the point where reexpression of this molecule could be induced. We, therefore, investigated the ability of different cytokines to mediate CD45RO to CD45RA reversion in freshly isolated CD8+ T cells that consisted of a large proportion of cells of the central memory type (data not shown).

We found that only IL-15 could generate CD45RA “revertants” from CD45RO+ EBV-specific T cells that were isolated from fresh PBMCs obtained from donors who had AIM at least 10 years previously (Figure 6). The freshly isolated EBV-specific CD8+ CD45RO+ population contained greater than 0.01% of cells that expressed CD45RA (Figure 6A, far left). These cells were cultured with either FCM (a recognized physiologic source of IFN-I19,20  which will be referred to as IFN-I for the rest of this manuscript), exogenous IL-2, IL-7, or IL-15 for 7 and 13 days, and the extent of cell cycling was determined by Ki67 expression (Figure 6A). As expected, there was no proliferation in the presence of IFN-I. In contrast, 55% of EBV tetramer+ cells that were cultured for 7 days in IL-15 were found to be Ki67 positive (Figure 6A). However, a lower percentage of EBV-specific CD8+ T cells exposed to IL-2 and IL-7 were found to be proliferating (12% and 14%, respectively) as determined by Ki67 expression (Figure 6A). The extent of cycling cells was reduced in all conditions by day 13 (Figure 6A). IL-15, but not IFN-I, IL-2, or IL-7, induced significant CD45RO to CD45RA reexpression in EBV tetramer+ T cells (Figure 6B), and by day 13 up to 16% of the cells showed complete loss of CD45RO. When examined on day 7, 27% and 13% of cells previously exposed to either IL-2 or IL-7, respectively, were CD45RA+. However, on day 13 very few CD45RA single-positive antigen-specific T cells were found in conditions in which either IL-2 (2%) or IL-7 (0%) were added (Figure 6B). By day 13, the number of cells found in conditions exposed to either IL-2 or IL-7 was much less than in those in which IL-15 was added (Figure 6B). This might be due to the fact that cells exposed to either IL-2 or IL-7 proliferated to a lesser degree, as demonstrated by a reduced Ki67 expression on day 7 (Figure 6A).

Figure 5.

Naturally occurring IFN-I cannot induce CD45RO to CD45RA reversion in antigen-specific T cells. CD45RA and CD45RO expression was determined in total CD8+ T cells (A) and EBV tetramer+ T cells gated on total CD8+ T cells (B) isolated from the same patient with AIM. CD45RA and CD45RO expression was also examined in freshly isolated antigen-specific T cells (C) and CD8+ tetramer+ T cells specifically activated and expanded in culture for 1 month (D). Cell-cycle progression was determined in activated CD8+ tetramer+ T cells by 7AAD staining (E). Activated virus-specific T cells were cultured with fibroblasts for 14 days at which point CD45RO expression (F) and 7AAD staining (G) were assessed by flow cytometric analysis. These results are representative of 4 separate experiments. (A-D, F) Numbers refer to the percentage of cells found in each quadrant.

Figure 5.

Naturally occurring IFN-I cannot induce CD45RO to CD45RA reversion in antigen-specific T cells. CD45RA and CD45RO expression was determined in total CD8+ T cells (A) and EBV tetramer+ T cells gated on total CD8+ T cells (B) isolated from the same patient with AIM. CD45RA and CD45RO expression was also examined in freshly isolated antigen-specific T cells (C) and CD8+ tetramer+ T cells specifically activated and expanded in culture for 1 month (D). Cell-cycle progression was determined in activated CD8+ tetramer+ T cells by 7AAD staining (E). Activated virus-specific T cells were cultured with fibroblasts for 14 days at which point CD45RO expression (F) and 7AAD staining (G) were assessed by flow cytometric analysis. These results are representative of 4 separate experiments. (A-D, F) Numbers refer to the percentage of cells found in each quadrant.

Close modal
Figure 6.

Generation of CD45RA revertants in CD8+ T cells by IL-15. CD8+ CD45RO+ T cells were negatively depleted from freshly isolated PBMCs and placed in culture with fresh medium supplemented by a single dose of either fibroblast-conditioned medium (FCM), IL-2, IL-7, or exogenous IL-15. The extent of proliferation in CD8+ tetramer+ T cells (A) was examined by using antibodies to Ki67 on days 0, 7, and 13. CD45R expression was also determined on days 0, 7, and 13 in CD8+ tetramer+ T-cell populations (B) and in total CD8+ T cells (B) exposed to either FCM or IL-15. (C) Western blot analysis of cell-cycle proteins (cyclin D2/D3, p27, and actin) in freshly isolated, resting CD8+ CD45RO+ memory T cells (lane 1). These CD8+ CD45RO+ memory T cells were subsequently exposed to a single dose of exogenous IL-15. The relative expression of cyclin D2/D3, p27, and actin was determined by Western blot analysis 7 (lane 2) and 13 days (lane 3) after exposure to IL-15. Cell-cycle–associated protein expression was determined by Western blot analysis in freshly isolated CD45RA+ CD27 revertant T cells (lane 4). These results are representative of 3 separate experiments.

Figure 6.

Generation of CD45RA revertants in CD8+ T cells by IL-15. CD8+ CD45RO+ T cells were negatively depleted from freshly isolated PBMCs and placed in culture with fresh medium supplemented by a single dose of either fibroblast-conditioned medium (FCM), IL-2, IL-7, or exogenous IL-15. The extent of proliferation in CD8+ tetramer+ T cells (A) was examined by using antibodies to Ki67 on days 0, 7, and 13. CD45R expression was also determined on days 0, 7, and 13 in CD8+ tetramer+ T-cell populations (B) and in total CD8+ T cells (B) exposed to either FCM or IL-15. (C) Western blot analysis of cell-cycle proteins (cyclin D2/D3, p27, and actin) in freshly isolated, resting CD8+ CD45RO+ memory T cells (lane 1). These CD8+ CD45RO+ memory T cells were subsequently exposed to a single dose of exogenous IL-15. The relative expression of cyclin D2/D3, p27, and actin was determined by Western blot analysis 7 (lane 2) and 13 days (lane 3) after exposure to IL-15. Cell-cycle–associated protein expression was determined by Western blot analysis in freshly isolated CD45RA+ CD27 revertant T cells (lane 4). These results are representative of 3 separate experiments.

Close modal

In these experiments, IFN-I was added at the beginning of culture and not replenished. In other similar experiments, where a dual chamber coculture system was used (confluent human embryonic lung fibroblasts and CD8+ T cells), no proliferation or CD45RA reexpression by freshly isolated CD8+CD45RO+ T cells was observed even after 3 weeks of coculture (data not shown). Finally, similar results were achieved in experiments in which total CD8+ T cells were examined (data not shown).

To rule out the possibility that CD45RA+ reexpression on EBV-specific CD8+ T cells was due to the expansion of small numbers (< 0.01%) of CD45RA+ contaminating cells, the CD45RO+ T cells were stained with CFSE prior to exposure to IL-15, and loss of staining was determined by FACS analysis 7 and 13 days later. We calculated the extent of expansion compared with the number of cell divisions that had occurred. These results showed that expansion of a contaminant population could not account for the frequency of EBV-specific CD8+ T cells observed (data not shown).

We next examined the effect of IL-15–induced “homeostatic” proliferation on cyclin expression in freshly isolated CD8+ CD45RO+ T cells (Table 1). These cells are not in cycle and express elevated levels of p27, no cyclin D3, and low levels of cyclin D2 (Figure 6C, lane 1). IL-15 induced these cells to proliferate and caused up-regulation of cyclin D3 after 7 days (Table 1). Cyclin D2 expression was also reduced, suggesting that this cyclin was not required for IL-15–induced homeostatic proliferation. At day 13, when the number of cells cycle in these cultures was much reduced (Figure 6A), cyclin D3 levels were reduced significantly (Figure 6C, lane 3). The p27 expression remained relatively constant throughout days 0 to 13 (Table 3). Finally, freshly isolated resting CD8+ CD45RA+ revertants express significant amounts of p27 as well as low levels of cyclins D2 and D3 (Table 3). Collectively, these results indicate that IL-15–induced homeostatic proliferation can induce CD45RA reexpression in EBV-specific CD8+ CD45RO+ T cells and that this proliferation is associated with a transient elevation in cyclin D3. Normalization ratios (based on optical densities) of antibody-positive protein bands versus β-actin controls were calculated to normalize the differences in β-actin expression between samples examined (Table 3). These ratios support the results described here.

Table 3.

Normalization ratios (based on optical densities) of antibody-positive protein bands versus β-actin controls



Days after IL-15 exposure

CD27-CD45RA+ revertant CD8+ T cells

0*
7
13
Cyclin D2  0.60   0.05   0.03   1.07  
Cyclin D3  0.22   1.00   0.74   0.53  
p27
 
0.54
 
0.57
 
0.60
 
0.52
 


Days after IL-15 exposure

CD27-CD45RA+ revertant CD8+ T cells

0*
7
13
Cyclin D2  0.60   0.05   0.03   1.07  
Cyclin D3  0.22   1.00   0.74   0.53  
p27
 
0.54
 
0.57
 
0.60
 
0.52
 
*

Prior to IL-15 exposure.

CD8+ CD45RA+ revertant T cells possess a higher threshold of activation but retain cytolytic function

The functional role of CD8+ T cells that reexpress CD45RA is not known. Previous reports suggest that, although these cells can be reactivated,7,8,12  the extent of proliferation elicited is less than that observed in other CD8 subsets.13  To address this we investigated the dose dependence for proliferation of freshly isolated naive (CD45RA+ CD27+), central memory (CD45RA CD27+), effector memory (CD45RA CD27), and revertant memory (CD45RA+ CD27) T cells after anti-CD3 and autologous APC stimulation (Figure 7A). Revertant T cells proliferated the least in response to T-cell receptor (TCR) stimulation, whereas the nave T-cell subset showed the best response (Figure 7A, left). However, the revertant CD8+ T cells exhibited the greatest cytotoxic activity (Figure 7A, right) even at low concentrations of anti-CD3 antibody, showing that they can mediate efficient effector function without proliferation. Furthermore, these cells were able to synthesize greater amounts of IFN-γ in response to stimulation compared with naive and central memory CD8+ T cells (Figure 7B). Finally, in Figure 7C we show that the CD8+ CD45RA+ revertant T cells isolated from healthy donors show almost complete loss of the costimulatory markers CD27 and CD28 that are crucial for stimulation, suggesting that this population has been functionally reprogrammed to mediate effector function instead of proliferation. It was not possible to carry out similar experiments on EBV-specific CD8+ T cells, as all 4 T-cell subsets, defined by the relative expression of CD27 and CD45RA, are not represented in these antigen-specific populations. In addition, there are too few EBV-specific CD8+ T cells in patients with chronic EBV infection to examine cytotoxic and proliferative function.

Figure 7.

Activation requirements of CD8+ CD45RA+ revertant T cells. CD8+ T-cell subsets were sorted using a MoFlo cell sorter on the basis of CD27 and CD45RA expression. (A) All CD8+ T-cell subsets were activated with a concentration range of anti-CD3 and irradiated autologous APCs. Proliferation was determined by tritiated thymidine incorporation 72 hours later (left). The cytotoxic potential of the same T-cell subsets was determined by assessing the relative expression of surface CD107 (right). ♦ indicates CD27+CD45RA+; ▪, CD27+CD45RA; ▴, CD27CD45RA; and ○, CD27CD45RA+. (B) T-cell subsets were also examined for INF-γ expression 18 hours after exposure to a concentration range of anti-CD3 plus autologous APCs. Finally, freshly isolated CD27CD45RA+ revertants (C, top) were stained for CD27 and CD28 expression (C, bottom). These results are representative of 3 separate experiments. Numbers refer to the percentage of cells in each quadrant.

Figure 7.

Activation requirements of CD8+ CD45RA+ revertant T cells. CD8+ T-cell subsets were sorted using a MoFlo cell sorter on the basis of CD27 and CD45RA expression. (A) All CD8+ T-cell subsets were activated with a concentration range of anti-CD3 and irradiated autologous APCs. Proliferation was determined by tritiated thymidine incorporation 72 hours later (left). The cytotoxic potential of the same T-cell subsets was determined by assessing the relative expression of surface CD107 (right). ♦ indicates CD27+CD45RA+; ▪, CD27+CD45RA; ▴, CD27CD45RA; and ○, CD27CD45RA+. (B) T-cell subsets were also examined for INF-γ expression 18 hours after exposure to a concentration range of anti-CD3 plus autologous APCs. Finally, freshly isolated CD27CD45RA+ revertants (C, top) were stained for CD27 and CD28 expression (C, bottom). These results are representative of 3 separate experiments. Numbers refer to the percentage of cells in each quadrant.

Close modal

Persistent infection imposes a significant strain on the proliferative capacity of CD8+ virus-specific T cells, and repeated episodes of T-cell activation may lead to the loss of specific T cells through telomere erosion and replicative senescence.5,6  Mechanisms that can keep memory T cells in a resting state, between episodes of stimulation, are, therefore, crucial to optimize their persistence in vivo. In addition, the ability to induce functional activity in memory T cells, without the need for proliferation, would minimize telomere erosion and prevent the loss of cells by replicative senescence. This would be particularly important for the maintenance of T-cell memory to chronic infective agents, such as EBV and CMV, whereby memory T cells may have to persist for more than 8 decades without significant input of new cells from the thymus.6  In this regard, the 3 key observations in this study are, first, that quiescence in activated EBV-specific CD8+ T cells can be induced by IFN-I. Second, CD45RA-reexpressing CD8+ T cells that accumulate during chronic viral infections and in elderly individuals represent an effector population that mediates potent functional activity without the need for proliferation. And finally, the reexpression of CD45RA by these EBV-specific CD8+ T cells can be induced during IL-15–mediated homeostatic proliferation.

We investigated the effects of IL-15 and IFN-I on EBV-specific T cells because these cytokines have been shown to be crucial for the maintenance of CD8+ T-cell memory in vivo; however, their mechanism of action is unclear.31-34  Both these groups of cytokines have been shown to prevent T-cell apoptosis in vitro and in vivo, which may be one way by which memory T cells are preserved.2,19,27,33,35,36  However, these cytokines may also have additional effects on memory CD8+ T-cell persistence through the induction of quiescence and CD45RA reexpression. Interestingly, these phenomena were found to be distinct; IFN-I induces quiescence, while IL-15 induces CD45RA reexpression. It must be noted that the timing and concentration levels of these 2 cytokines are likely to be crucial as to the effect on local T cells. IFN-α–induced IL-15 has been shown to be important for driving immune responses,30  whereas it is an established fact that IFN-α exhibits potent antiviral and immunomodulatory roles at the start of an immune response.33  However, once local supplies of γ-chain cytokines and antigen-presenting cells become exhausted, it is probable that IFN-I, secreted by stromal cells in the microenvironment,6,28  can promote the survival of any remaining activated T cells. We (data not shown) and others13  have suggested that IL-15 is not capable of generating CD45RA revertants from TCR-activated T cells. It is likely, therefore, that IL-15–mediated reversion occurs in noninflamed tissues as part of the homeostatic maintenance of CD8+ memory T cells in persistent viral infection.

We examined cyclin expression in freshly isolated virus-specific T cells to determine the molecular signatures of quiescence and activation in these cells. Recent reports in mice showed that resting CD8+ memory T cells possess high cyclin D3,37,38  which may permit a more rapid response to recall antigen.38  In contrast, we found high cyclin D2 expression and the complete absence of cyclin D3 in resting human CD8+CD45RO+ memory T cells. These discrepancies may reflect species-specific differences.39  EBV-specific CD8+ T cells that were freshly isolated from patients with AIM or activated in vitro showed very similar cyclin signatures with up-regulated cyclin D2 and D3 but down-regulation of p27. The rescue of activated EBV-specific T cells from apoptosis with either IFN-I or IL-15 induced different cyclin profiles, the addition of IFN-I down-regulated cyclin D3, whereas IL-15 induced cyclin D3 expression. While we cannot rule out the possibility that the increase in p27 by IFN-I in our experiments is simply due to removal of the stimulus, it is likely that there is also a component of active p27 induction by these cytokines.21,29,40 

Primed CD8+ T cells that reexpress CD45RA are found after a variety of different viral infections, including EBV, CMV, and HIV in humans 4,7,8,13,24,41-43 , and a similar subset is also found in animals.44  Our data suggest that the induction of CD45RA reexpression in CD8+ T cells is part of a functional reprogramming of effector memory cells away from proliferation that also includes the loss of both CD27 and CD28 costimulatory molecules. The loss of these costimulatory molecules also occurs in EBV-specific effector memory CD8+ T cells that do not reexpress CD45RA.43,45-47  This suggests that loss of costimulatory molecule expression alone cannot account for the poor proliferative activity of this population. Nevertheless, the potent cytotoxic activity and capacity for IFN-γ secretion by these cells,8,13,24  together with their capacity to migrate to nonlymphoid tissue,45  show that they are ideally suited for a role in immunosurveillance. Signals associated with the generation of CD45RA revertants by IL-15 in virus-specific CD8+ T cells are unclear; however, since the IL-2R shares both the β- and γ-chains with the IL-15R48  but does not induce reversion (data not shown), IL-15R α-chain–specific signals are likely to be involved.

In summary, memory T cells have to be maintained for many decades in humans, and cells that are specific for persistent infective agents will be repeatedly stimulated during life-long infection. This predisposes these populations to loss by telomere-dependent replicative senescence.5,6  Quiescence-inducing factors such as IFN-I and those that induce CD45RA reexpression like IL-15 are mediators that may, therefore, be crucial for the long-term maintenance of these cells. It is not clear whether there is an equivalent murine CD8+ memory T-cell population that is polarized away from proliferation but toward effector function. However, the much shorter mouse compared with human life span suggests that these cells would be more important in the latter species. A closer understanding of the timing and location of endogenous production of these cytokines in vivo will give a clearer rationale for vaccine therapy, especially in elderly individuals.

Prepublished online as Blood First Edition Paper, March 29, 2005; DOI 10.1182/blood-2004-11-4469.

Supported by a grant from Dermatrust, the Edward Jenner Institute for Vaccine Research (P.J.D.), the Biotechnology and Biological Sciences Research Council (BBSRC) (L.B. and M.V.D.S.), the Arthritis Research Campaign (ARC) (M.S.), and the Leukemia Research Fund (E.W.-F.L. and S.F.dM.).

P.J.D. designed, performed, and analyzed the research and wrote the paper; L.B., J.M.F., M.L., and M.V.D.S. performed the research; S.F.dM. performed the research and contributed analytical tools; M.H.A.R. designed the research; E.W.-F.L. designed the research and contributed vital new reagents and analytical tools; M.S. designed the research and analyzed the data; A.N.A. designed and analyzed the research and wrote the paper.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

We thank Professor Peter C.L. Beverley for valuable discussions and Mr Andrew Worth for excellent assistance with cell sorting.

1
Zinkernagel RM, Bachmann MF, Kundig TM, et al. On immunological memory.
Annu Rev Immunol.
1996
;
14
:
333
-367.
2
Akbar AN, Salmon M. Cellular environments and apoptosis: tissue microenvironments control activated T cell death.
Immunol Today.
1997
;
18
:
72
-76.
3
Rickinson AB, Moss DJ. Human cytotoxic T lymphocyte responses to Epstein-Barr virus infection.
Annu Rev Immunol.
1997
;
15
:
405
-431.
4
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.
5
Hodes RJ, Hathcock KS, Weng NP. Telomeres in T and B cells.
Nat Rev Immunol.
2002
;
2
:
699
-706.
6
Akbar AN, Beverley PC, Salmon M. Will telomere erosion lead to a loss of T-cell memory?
Nat Rev Immunol.
2004
;
4
:
737
-743.
7
Wills MR, Carmichael AJ, Weekes MP, et al. Human virus-specific CD8+ CTL clones revert from CD45ROhigh to CD45RAhigh in vivo: CD45RAhighCD8+ T cells compromise both naive and memory cells.
J Immunol.
1999
;
162
:
7080
-7087.
8
Dunne PJ, Faint JM, Gudgeon NH, et al. Epstein-Barr virus-specific CD8+ T cells that re-express CD45RA are apoptosis-resistant memory cells that retain replicative potential.
Blood.
2002
;
100
:
933
-940.
9
Grubeck-Loebenstein B, Wick G. The aging of the immune system.
Adv Immunol.
2002
;
80
:
243
-284.
10
Champagne P, Ogg GS, King AS, et al. Skewed maturation of memory HIV-specific CD8 T lymphocytes.
Nature.
2001
;
410
:
106
-111.
11
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.
12
van Lier RA, ten Berge IJ, Gamadia LE. Human CD8(+) T-cell differentiation in response to viruses.
Nat Rev Immunol.
2003
;
3
:
931
-939.
13
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
-4266.
14
Wallace DL, Zhang Y, Ghattas H, et al. Direct measurement of T cell subset kinetics in vivo in elderly men and women.
J Immunol.
2004
;
173
:
1787
-1794.
15
Balomenos D, Martinez-A C. Cell-cycle regulation in immunity, tolerance and autoimmunity.
Immunol Today.
2000
;
21
:
551
-555.
16
Walworth NC. Cell-cycle checkpoint kinases: checking in on the cell cycle.
Curr Opin Cell Biol.
2000
;
12
:
697
-704.
17
Yusuf I, Fruman DA. Regulation of quiescence in lymphocytes.
Trends Immunol.
2003
;
24
:
380
-386.
18
Zhang S, Lawless VA, Kaplan MH. Cytokine-stimulated T lymphocyte proliferation is regulated by p27Kip1.
J Immunol.
2000
;
165
:
6270
-5277.
19
Pilling D, Akbar AN, Girdlestone J, et al. Interferon-β mediates stromal cell rescue of T cells from apoptosis.
Eur J Immunol.
1999
;
29
:
1041
-1050.
20
Lombardi G, Dunne PJ, Scheel-Toellner D, et al. Type I IFN maintains the survival of anergic CD4+ T cells.
J Immunol.
2000
;
165
:
3782
-3789.
21
Grander D, Sangfelt O, Erickson S. How does interferon exert its cell growth inhibitory effect?
Eur J Haematol.
1997
;
59
:
129
-135.
22
De Maeyer E, De Maeyer-Guignard J. Type I interferon.
Int Rev Immunol.
1998
;
17
:
53
-73.
23
Plunkett FJ, Soares MVD, Annels NE, et al. The flow cytometric analysis of telomere length in antigen-specific CD8+ T cells during acute Epstein Barr virus infection.
Blood.
2001
;
97
:
1
-8.
24
Hislop AD, Gudgeon NH, Callan MFC, et al. EBV-specific CD8+ T cell memory: relationships between epitope specificity, cell phenotype, and immediate effector function.
J Immunol.
2001
;
167
:
2019
-2029.
25
Reed JR, Vukmanovic-Stejic M, Fletcher JM, et al. Telomere erosion in memory T cells induced by telomerase inhibition at the site of antigenic challenge in vivo.
J Exp Med.
2004
;
199
:
1433
-1443.
26
Betts MR, Brenchley JM, Price DA, et al. Sensitive and viable identification of antigen-specific CD8+ T cells by a flow cytometric assay for degranulation.
J Immunol Methods.
2003
;
281
:
65
-78.
27
Soares MV, Plunkett FJ, Verbeke CS, et al. Integration of apoptosis and telomere erosion in virus-specific CD8+ T cells from blood and tonsils during primary infection.
Blood.
2004
;
103
:
162
-167.
28
Akbar AN, Borthwick NJ, Salmon M, et al. The significance of low bcl-2 expression by CD45RO T cells in normal individuals and patients with acute viral infections. The role of apoptosis in T cell memory.
J Exp Med.
1993
;
178
:
427
-438.
29
Moro A, Santos A, Arana MJ, Perea SE. Activation of the human p27Kip1 promoter by IFNα 2b.
Biochem Biophys Res Commun.
2000
;
269
:
31
-34.
30
Gombert W, Borthwick NJ, Wallace DL, et al. Fibroblasts prevent apoptosis of IL-2-deprived T cells without inducing proliferation: a selective effect Bcl-xL expression.
Immunology.
1996
;
89
:
397
-404.
31
Sprent J, Tough DF. T cell death and memory.
Science.
2001
;
293
:
245
-248.
32
Sprent J, Surh CD. Generation and maintenance of memory T cells.
Curr Opin Immunol.
2001
;
13
:
248
-254.
33
Gourley TS, Wherry EJ, Masopust D, Ahmed R. Generation and maintenance of immunological memory.
Semin Immunol.
2004
;
16
:
323
-333.
34
Schluns KS, Lefrancois L. Cytokine control of memory T-cell development and survival.
Nat Rev Immunol.
2003
;
3
:
269
-279.
35
Marrack P, Kappler J, Mitchell T. Type I interferons keep activated T cells alive.
J Exp Med.
1999
;
189
:
521
-529.
36
McHeyzer-Williams M, McHeyzer-Williams L, Panus J, et al. Helper T-cell-regulated B-cell immunity.
Microbes Infect.
2003
;
5
:
205
-212.
37
Bartkova J, Lukas J, Strauss M, Bartek J. Cyclin D3: requirement for G1/S transition and high abundance in quiescent tissues suggest a dual role in proliferation and differentiation.
Oncogene.
1998
;
17
:
1027
-1037.
38
Veiga-Fernandes H, Rocha B. High expression of active CDK6 in the cytoplasm of CD8 memory cells favors rapid division.
Nat Immunol.
2004
;
5
:
31
-37.
39
Mestas J, Hughes CC. Of mice and not men: differences between mouse and human immunology.
J Immunol.
2004
;
172
:
2731
-2738.
40
Erickson S, Sangfelt O, Castro J, et al. Interferon-alpha inhibits proliferation in human T lymphocytes by abrogation of interleukin-2-induced changes in cell-cycle-regulatory proteins.
Cell Growth Differ.
1999
;
10
:
575
-582.
41
Callan MFC, Tan L, Annels NE, et al. Direct visualization of antigen-specific CD8+ T cells during the primary immune responses to Epstein-Barr virus in vivo.
J Exp Med.
1998
;
187
:
1395
-1402.
42
Rufer N, Zippelius A, Batard P, et al. Ex vivo characterization of human CD8+ T subsets with distinct replicative history and partial effector functions.
Blood.
2003
;
102
:
1779
-1787.
43
Appay V, Dunbar PR, Callan MFC, et al. Memory CD8+ T cells vary in differentiation phenotype in different persistent virus infections.
Nat Med.
2002
;
8
:
379
-385.
44
Bell EB, Hayes S, McDonagh M, et al. Both CD45Rlow and CD45Rhigh `revertant' CD4 memory T cells provide help for memory B cells.
Eur J Immunol.
2001
;
31
:
1685
-1695.
45
Faint JM, Annels NE, Curnow SJ, et al. Memory T cells constitute a subset of the human CD8+CD45RA+ pool with distinct phenotypic and migratory characteristics.
J Immunol.
2001
;
167
:
212
-220.
46
van Baarle D, Kostense S, van Oers MH, Hamann D, Miedema F. Failing immune control as a result of impaired CD8+ T-cell maturation: CD27 might provide a clue.
Trends Immunol.
2002
;
23
:
586
-591.
47
Klebanoff CA, Finkelstein SE, Surman DR, et al. IL-15 enhances the in vivo antitumor activity of tumor-reactive CD8+ T cells.
Proc Natl Acad Sci U S A.
2004
;
101
:
1969
-1974.
48
Waldmann TA. The IL-2/IL-15 receptor systems: targets for immunotherapy.
J Clin Immunol.
2002
;
22
:
51
-56.
Sign in via your Institution