Human CD8+ memory- and effector-type T cells are poorly defined. We show here that, next to a naive compartment, two discrete primed subpopulations can be found within the circulating human CD8+ T cell subset. First, CD45RACD45R0+ cells are reminiscent of memory-type T cells in that they express elevated levels of CD95 (Fas) and the integrin family members CD11a, CD18, CD29, CD49d, and CD49e, compared to naive CD8+ T cells, and are able to secrete not only interleukin (IL) 2 but also interferon γ, tumor necrosis factor α, and IL-4. This subset does not exert cytolytic activity without prior in vitro stimulation but does contain virus-specific cytotoxic T lymphocyte (CTL) precursors. A second primed population is characterized by CD45RA expression with concomitant absence of expression of the costimulatory molecules CD27 and CD28. The CD8+CD45RA+CD27 population contains T cells expressing high levels of CD11a, CD11b, CD18, and CD49d, whereas CD62L (L-selectin) is not expressed. These T cells do not secrete IL-2 or -4 but can produce IFN-γ and TNF-α. In accordance with this finding, cells contained within this subpopulation depend for proliferation on exogenous growth factors such as IL-2 and -15. Interestingly, CD8+CD45RA+CD27 cells parallel effector CTLs, as they abundantly express Fas-ligand mRNA, contain perforin and granzyme B, and have high cytolytic activity without in vitro prestimulation. Based on both phenotypic and functional properties, we conclude that memory- and effector-type T cells can be separated as distinct entities within the human CD8+ T cell subset.

Recognition of antigen by the immune system evokes a coordinate number of changes in lymphocytes and lymphocyte subsets that allow the system (a) to eliminate or neutralize potential harmful agents and (b) to respond more rapidly and appropriately after renewed antigen encounter, a process referred to as immunological memory. Within the T cell compartment, unprimed (or naive) T cells (i.e., cells which have not yet encountered antigen), effector T cells (i.e., cells with specialized functions such as cytolysis), and memory T cells can be discerned. Functionally, memory T cells have a number of characteristics that distinguish them from unprimed cells. Not only do they respond efficiently to recall antigens, but, compared to naive cells, memory T cells also have less stringent requirements for activation and have the potential to secrete a more extensive set of cytokines (14). In addition, memory and naive T cells differ in the expression of several cell surface antigens (5, 6), although it is questioned whether these markers can be looked upon as true memory markers or if they reflect cellular activation (7, 8).

Recent studies by Zimmermann et al. (9) have documented phenotypic distinctions between naive, effector, and memory CD8+ T cells in a murine model of lymphocytic choriomeningitis virus infection and an adoptive transfer system with CD8+ T cells from TCR transgenic mice. Their data confirmed previous reports (1014) by demonstrating that compared to naive T cells CTL effectors have strongly downregulated the lymph node homing receptor CD62L (L-selectin) while expression of CD44, CD11a/CD18, CD11b, and CD49d is enhanced. In contrast to effector T cells, memory T cells are heterogenous with respect to CD62L expression, have no CD11b on their surface, and have an intermediate CD49d expression. Finally, CD45 isoform expression could not be used as a reliable marker for memory-type T cells (9).

In contrast to the murine T cell subsets, description of human CD8+ naive, effector, and memory cells is still rather fragmentary. Cytolytic function of CD8+ cells has been linked to cells with a CD28, a CD11b+, and/or a CD57+ phenotype (11, 15, 16). Moreover, memory-type CD8+ CD45RACD45R0+ T cell subsets that contain enhanced frequencies of antigen-reactive precursor cells (17), have low stringent activation requirements, and have an ability to secrete, apart from IL-2, IFN-γ and TNF-α (18), have been previously described. However, discrimination between naive and memory cells, based solely on CD45 isoform expression, has been proven unreliable, since the CD45RA+ population contains cells that share a number of phenotypic features with primed cells, such as low CD62L (19) and high CD11a/CD18 (20) expression. Moreover, part of the CD45RA+ cells have downregulated CD27 (21), a feature considered to result from persistent antigenic stimulation in vivo (22, 23).

In this study we used simultaneous staining with CD45RA and CD27 mAbs to separate functionally distinct subpopulations of human CD8+ T cells. Our observations demonstrate that, within the human CD8+ compartment, in accordance with data obtained in experimental animal models, naive, memory- and effector-type cells can be discriminated with coherent phenotypic and functional properties. This type of CD8+ T cell subset analysis could prove useful in monitoring the immune system in several clinical situations.

Reagents.

The mAbs CLB-T11.1/1, CLB-T11.2/1, CLB-HIK27 (all CD2), CLB-CD3/3 CLB-CD3/4.1, CLB-CD4/1, CLB-CD8/1, CLB-CD14/1, CLB-FcR gran1 (CD16), CLB-CD19/1, CLB-CD27/1, CLB-CD28, CLB-LFA-1/1 (CD18), CLB-CD49d (VLA-4), and CLB-CD49e (VLA-5), and FITC-conjugated goat anti–mouse Ig, were all produced at the Central Laboratory of the Netherlands Red Cross Blood Transfusion Service (CLB; Amsterdam, The Netherlands). CD95 mAb was provided by Dr. Yonehara (Pharmaceutical Basic Research, Tokyo, Japan) and CD58 mAb (TS2/9) was a gift from Dr. T.A. Springer (Center for Blood Research, Boston, MA). CD11b (OKM-1) mAb was purchased from Ortho Diagnostic Systems (Beerse, Belgium). FITC-conjugated–CD62L (Leu-8), –CD57, –CD16, –TCR-α/β, and –CD8 mAbs (Leu2a), and PE-conjugated– and PerCP-labeled CD8 mAb were from Becton Dickinson (San Jose, CA). CD29 (4B4) and PE-conjugated CD45RA mAbs (2H4) were obtained from Coulter Immunology (Hialeah, FL). FITC-labeled CD45RO mAb (UCHL1) was purchased from DAKO (Glostrup, Denmark). Streptavidin red 670 was obtained from GIBCO BRL (Gaithersburg, MD).

Biotinylated CLB-IL-4/1 mAb (anti–human IL-4, mouse IgG1; reference 24) was provided by Dr. T. van der Pouw-Kraan (CLB) and biotinylated MD-1 mAb (anti–human IFN-γ, mouse IgG1) was obtained from Dr. P. van der Meide (Netherlands Organisation for Applied Sciences TNO, Rijswijk, The Netherlands). Biotinylated anti–human IL-2, FITC-conjugated anti– human TNF-α, and FITC-conjugated antiperforin mAbs were purchased from Hölzel Diagnostika (Köln, Germany). Antigranzyme B mAb, provided by Dr. C.E. Hack (CLB), was obtained after immunization of BALB/c mice with native granzyme B.

The following recombinant human cytokines were used: IL-4 and IFN-γ (Genzyme Corp., Cambridge, MA), IL-15 (PeproTech, London, UK), IL-2 (gift of Sandoz, Vienna, Austria), IL-6 (provided by Professor L. Aarden, CLB), IL-10 (gift of Dr. J. de Vries, DNAX, Palo Alto, CA), and IL-12 (donated by Dr. S. Wolf, Genetic Institute, Cambridge, MA).

Cell Preparation and Flow Cytometry.

Human PBMCs were isolated from buffy coats of healthy donors by Ficoll-Paque density centrifugation (Pharmacia Biotech AB, Uppsala, Sweden). For phenotypic analysis (Table 1), CD8+ cells (>95% CD8+ CD3+TCR-α/β+CD16) were purified by incubating PBMCs with saturating amounts of CD4, CD19, CD16, and CD14 mAbs followed by depletion of positive cells with Dynal beads (Dynal, Oslo, Norway) as previously described (18). Triple color immunofluorescence analysis was performed as previously described (23). In brief, purified CD8+ T cells were incubated with an unlabeled mAb followed by FITC-conjugated goat anti–mouse Ig staining. After blocking free binding sites on the goat anti–mouse conjugate with 10% normal mouse serum, cells were incubated with PE-conjugated CD45RA and biotinylated CD27 mAbs. The latter was then stained with Streptavidin red 670.

For subset purification, CD8+ T cells were prepared by positive enrichment using the MACS system (Miltenyi Biotec, Bergisch-Gladbach, Germany). In brief, PBMCs (107 cells/80 μl incubation buffer; PBS/2% FCS/5 mM EDTA) were stained with CD8 microbeads (Miltenyi Biotec; 20 μl/107 positive cells) for 15 min at 8°C. After one washing, step cells were resuspended in incubation buffer (107 cells/400 μl) and enrichment was performed with BS columns (capacity: 108 cells) and the VarioMACS magnet according to manufacturer's instructions. The resulting CD8+ T cells were >98% CD8+CD3+TCR-α/β+CD16 as determined by immunofluorescence analysis with directly labeled mAb. Next, cells were stained with PE-conjugated CD45RA and FITC-conjugated CD27 and sorted into CD45RA+CD27+, CD45RA+ CD27, and CD45RACD27+ populations (purity: >98%) on a FACStar®.

Detection of Fas-ligand mRNA.

Total RNA was prepared (TRIzol Reagent; Life Technologies, Paisley, UK) from 2.5 × 106 sorted T cell subsets (see above). Single-stranded cDNA was synthesized from RNA in a 20-μl reaction mixture containing 500 ng oligo(dT)12–18 and 200 U reverse transcriptase (RT).1 1 μl of the reaction mixture was diluted with 11.5 μl of PCR buffer containing 2 mM MgCl2 and 100 pmol of forward and reverse oligonucleotides. Two PCR reactions were set up for each cDNA, corresponding to Fas ligand (forward: 5′-GGGTCGACGGGATGTTTCAGCTCTTCCACCTAC - 3′ ; reverse: 5′-GCTCTAGAACATTCCTCGGTGCCTGTAAC-3′; reference 25) and HPRT (hypoxanthine-guanine phosphoribosyltransferase; forward: 5′-TATGGACAGGACTGAACGTCTTGC-3′; reverse: 5′-GACACAAACATGATTCAAATCCCTGA-3′; reference 26). PCR products were resolved on a 1% agarose gel containing ethidium bromide.

Flow Cytometric Detection of Cytokine Production and Intracellular Staining for Perforin and Granzyme B.

Flow cytometric measurement of cytokine production was performed as previously described (27, 28). In brief, 106 cells/ml were stimulated for 4 h (IL-4, IFN-γ, and TNF-α) or 8 h (IL-2) with 1 ng/ml PMA and 1 μM ionomycin in the presence of the protein-secretion inhibitor monensin (1 μM; all from Sigma Chemical Co., St. Louis, MO). This short-term stimulation did not alter the membrane phenotype with respect to CD45 or CD27 expression. After cell surface staining with PE-conjugated CD45RA and FITC-conjugated CD27 cells were washed twice with cold PBS and fixated with PBS/4% paraformaldehyde (at 4°C for 5 min). Fixation was followed by permeabilization with PBS/0.1% saponin (Sigma Chemical Co.)/10% human pooled serum (at 4°C for 10 min). For all subsequent incubation and washing steps, PBS/0.1% saponin/0.5% BSA was used. Staining of the cytoplasm with biotinylated cytokine mAbs (IL-2, IL-4, and IFN-γ; all 5 μg/ml) was followed by incubation with Streptavidin red 670 (both at 4°C for 20 min). Biotinylated or FITC-conjugated mouse IgG1 control mAbs (DAKO) were used as negative controls. Data acquisition was performed on a FACScan® and analysis was done using PC-Lysis software.

Intracellular content of perforin and granzyme B was measured in freshly isolated CD8+ cells without previous stimulation. A permeabilization and staining protocol identical to that described above for the cytokine analysis was used.

Cell Culture and Activation.

All cell culture experiments were performed in IMDM supplemented with 10% FCS and antibiotics. Sorted CD8+ T cell subsets (5 × 104 cells/well) were stimulated with a combination of three CD2 mAbs (CLB-T11.1/1, CLB-T11.2/1, and CLB-HIK27, all 5 μg/ml) in the presence or absence of the following recombinant human cytokines: IL-2 (50 U/ml), IL-4 (10 ng/ml), IL-6 (100 U/ml), IL-10 (100 U/ml), IL-12 (1 ng/ml), IL-15 (10 ng/ml), and IFN-γ (100 U/ml). Proliferation was measured on day 4 by adding 0.2 μCi/well of [3H]thymidine (Amersham, Buckinghamshire, UK) during the last 4 h of culture. All cultures were set up in triplicate.

To measure cytokine secretion, triplicate cultures of sorted cells (2.5 × 104) were stimulated with PMA (100 ng/ml) and ionomycin (1 μM). Supernatant was taken after 24 h for detection of IL-2 and on day 4 for measurement of IFN-γ, TNF-α, and IL-4 content. In our experience, the indicated time points have been proven optimal. IFN-γ, TNF-α, and IL-4 were detected with specific ELISAs (CLB). IL-2 production was measured in a bioassay using IL-2–dependent mouse CTLL cells (29).

Cytotoxicity Assay.

CTL activity was determined in a CD3 mAb–mediated cytotoxicity assay as previously described (30). In brief, FcR-bearing P815 target cells were radiolabeled with Na51CrO4 (Amersham) for 30 min at 37°C. Purified CD8+ T cell subsets were incubated with P815 target cells at varying effector/ target ratios in the presence or absence of 5 μg/ml CD3 mAb (CLB-CD3/4.1). After a 4 h incubation period at 37°C, the supernatants of triplicate cultures were collected and counted in a gamma counter. Specific cytotoxicity was determined according to the formula: percentage of specific lysis = 100 × [(cpm experimental release − cpm spontaneous release)/(cpm maximal release − cpm spontaneous release)]. The standard error of the mean percentage lysis did not exceed 5%.

Determination of HIV-1–derived Peptide-specific CTL Precursor Frequency.

HIV-1–specific CTL precursors (CTLp) were expanded in vitro by Ag-specific stimulation and frequencies of RT derived peptide aa 244-252 (IVLPEKDSW)–specific CTLp were determined as previously described (31, 32). In brief, cryopreserved PBMCs from an HIV-1 seropositive individual (L090; for case report see reference 32) were thawed and sorted into CD8+ CD45RA+CD27+ and CD8+CD45RACD27+ subsets. Sorted cells were cocultivated with stimulator cells (paraformaldehyde fixated autologous EBV-transformed B lymphoblastoid cell lines [B-LCL] infected with five multiplicity of infection of recombinant vaccinia virus expressing HIV-1Lai RT) and irradiated feeder cells (autologous PBMCs; references 31, 32). At days 2 and 9, microcultures were fed with rIL-2 (10 U/ml Proleukin; provided by Dr. R. Rombouts, Chiron Benelux B.V., Amsterdam, The Netherlands). Cultures were restimulated at day 7 by addition of fixated autologous stimulator cells in the presence of rIL-2 (10 U/ml). On day 15, wells were split and tested for cytotoxicity on 51Cr-labeled autologous B-LCL that were preincubated for 30 min at room temperature with synthetic peptide at a final concentration of 5 μg/ ml. Lysis of target cells was measured as described above. Wells were considered positive when 51Cr-release exceeded 10% of specific lysis. Frequency estimates were made using the “single-hit Poisson model” described by Strijbosch et al. (33).

Helper and Suppressor Activity for Ig Production.

CD8+ T cells and CD8+ subsets were prepared as described above. The CD8 fraction obtained after purification of CD8+ cells was either depleted of monocytes, NK cells, the remaining CD8+ T cells, and B cells with CD14, CD16, CD8, and CD19 mAbs and magnetic beads (Dynal) to generate CD4+ T cells (>95% CD4+) or else depleted of monocytes, NK cells, and T cells with CD14, CD16, and CD3 mAbs to prepare B cells (>90% CD19+). To determine helper capacity, cells of the different T cell subsets (5 × 104/well) were cultured with autologous B cells (5 × 104/well) in 96-well flat-bottomed microtiter plates (Greiner, Nürnberg, Germany) coated with 5 μg/ml CD3 mAb (CLB-CD3/3). Suppressor activity was determined by adding cells of the different CD8+ T cell subsets (5 × 104/well) to a coculture of autologous CD4+ T and B cells. All cultures were set up in triplicate and supernatants were harvested after 14 d and tested for Ig content. IgM and IgG production was measured by ELISA.

Phenotypic Heterogeneity of the CD8+ T Cell Subset.

As has been previously noted (21), in contrast to CD4+CD27 T lymphocytes, which are exclusively CD45RA, within the circulating CD8+ compartment CD27 T cells can be found within both CD45RA and CD45RA+ fractions (Fig. 1). These CD8+CD45RA+CD27 cells are, like CD8+ CD45RACD45R0+ cells, absent from cord blood (Fig. 1). Therefore, we hypothesize that in response to antigen two separate primed CD8+ populations may develop in humans: (a) a population of CD45RA T cells, and (b) a population of CD45RA+CD27 T cells. This study was performed to analyze whether these phenotypes discriminate between functionally distinct CD8+ subsets.

In a population of healthy adult donors (n = 30), the distribution of the CD8+ T cell subsets defined by simultaneous CD45RA and CD27 staining was as follows (for definition of the subsets see Fig. 1): (1) CD45RA+CD27+ cells 55 ± 17% (range 28–83%); (2) CD45RACD27+ cells 25 ± 11% (range 9–48%); (3) CD45RA+CD27 cells 13 ± 13% (range 1–50%); and (4) CD45RACD27 cells 4 ± 3% (range 0–15%).

To further define the cell surface phenotype of these subsets, CD8+ T cells were purified from peripheral blood and analyzed in triple color immunofluorescence. As expected on the basis of the observation that these were the only cells present in cord blood, CD45RA+CD27+ cells showed characteristics of unprimed cells since they were CD62L+ and had low CD49d, CD49e, CD29, CD11a/ CD18, and CD58 expression. The majority of cells were CD28+ and did not express CD45R0 (Table 1 and data not shown). The CD45RA population had features of antigen-primed cells. CD62L was downregulated, and CD49e, CD29, and CD58 were upregulated when compared to the unprimed population. The main difference between CD27+ and CD27 cells within the CD45RA population was the expression of CD28. Although the majority of CD27+ cells expressed the antigen, only half of the CD27 cells were CD28+.

Although CD45R0 was absent from CD45RA+CD27 cells, these T cells had a number of features compatible with a primed phenotype. CD62L was found to be downregulated, whereas CD49d, CD49e, CD29, CD11a/CD18, and CD58 were highly expressed. In comparison to the CD45RA population, expression levels of CD49d and CD11a/CD18 were even higher on CD45RA+CD27 cells. Remarkably, CD11b and CD57 were present on >50% of the cells and the majority did not express CD28.

Fas (CD95) was not expressed on CD8+CD45RA+ CD27+ lymphocytes but could be detected on the majority of CD45RA+CD27 T cells. It should be noted that the antigen density on the latter population was markedly lower than on CD8+CD45RA T cells (Table 1 and Fig. 2,A). Interestingly, Fas ligand mRNA was not present in CD8+CD45RA+CD27+ T cells but was strongly expressed in CD8+CD45RA+CD27 lymphocytes (Fig. 2,B, lanes B and C). In contrast, only trace amounts of Fas ligand mRNA were detected in CD8+CD45RACD27+ cells (Fig. 2 B, lane D).

Finally, it should be noted that a CD8+CD27+ population expressing low levels of CD45RA can be found in most donors (Fig. 1). Analogous to similar cells within the CD4+ T cell compartment (34), these cells coexpress CD45R0 and were found to have an intermediate membrane phenotype between CD45RA+CD27+ and CD45RA CD27+ T cells (data not shown).

Cytokine Production Capacity of CD8+ T Cell Subsets.

In contrast to naive T cells, which primarily secrete IL-2, the ability to produce a variety of cytokines is a typical feature of primed T cells (35). Cytokine production capacity of CD8+ T cell subsets was measured after stimulation for 4 h with PMA and ionomycin at a single cell level (Fig. 3 and Table 2). CD8+CD45RA+CD27+ cells paralleled naive CD4+ cells in that IL-2 was the main cytokine produced by this subset. In agreement with what would be expected from primed cells, CD8+CD45RA cells were able to secrete IL-2, IL-4, IFN-γ, and TNF-α. IL-4+ cells were exclusively found within this population with both CD27+ and CD27 cells contributing to the total IL-4 secretion. Also with respect to cytokine expression, the CD45RA+ CD27 subset showed characteristics of antigen-experienced cells, in that a high percentage of IFN-γ and TNF-α producers could be detected. Remarkably, in contrast to CD45RA cells, neither IL-2 nor IL-4 production was found. Since IL-2 production in CD45RA+ cells has been described to reach its maximum later than in CD45RA cells (36), we also analyzed 8-h stimulated cells. Indeed, the frequency of IL-2 producers increased in the CD45RA+ CD27+ subset. However, also at this time point, CD45RA+ CD27 cells did not produce any IL-2. From Fig. 3 it should be noted that, with respect to the cytokine secretion pattern, cells with a CD27dull expression behave as CD27 T cells in this assay. Furthermore, expression of surface markers on these cells was also comparable to CD27 cells (data not shown).

The results obtained by intracellular cytokine staining were confirmed by measuring the cytokine content of the supernatant after 4 d of culture in the presence of PMA and ionomycin (Table 3). Again, CD45RA+CD27+ T cells mainly produced IL-2. CD45RA cells secreted all measured cytokines, whereas CD45RA+CD27 cells were only capable of IFN-γ and TNF-α production.

To test if the distinction in cytokine production profiles between the two primed CD8+ T cell subpopulations was reflected in their proliferation capacities, three fractions were sorted from purified CD8+ T cells, i.e., CD45RA+CD27+, CD45RA+CD27, and CD45RACD27+ cells, and stimulated with a combination of CD2 mAbs supplemented with various cytokines (Fig. 4). The CD45RACD27 population could not be analyzed because of the low number of these cells in most donors (see above). Culture of the sorted cells with the indicated cytokines alone did not result in proliferation (data not shown). In accordance with a previous report (18) on stringent activation requirements of unprimed CD8+ T cells, CD45RA+CD27+ cells were not able to proliferate in response to CD2 mAbs alone, but addition of IL-2, -15, -4, and, to a lesser extent, -12 resulted in a vigorous proliferative response, whereas IL-6 and -10 and IFN-γ had no effect on proliferation. In contrast, CD2 mAbs alone were sufficient to induce proliferation of CD45RACD27+ cells. IL-2, -15, and -12 further increased proliferation, but IL-4, -6, and -10, and IFN-γ only marginally influenced the CD2-induced response. Proliferation of CD45RA+CD27 cells was only observed in the presence of either IL-2 or -15. However, the magnitude of the response was lower than that of the other two populations.

CD45RA+CD27, but not CD45RACD27+ T Cells, Have Cytolytic Activity without Previous In Vitro Stimulation.

Our data thus far have shown that CD45RA+ CD27 cells possess a number of phenotypic characteristics that have been associated with murine (9) and human (11, 15, 16, 37) cytolytic T cells and that these cells secrete IFN-γ and TNF-α, two lymphokines known to play a role in controlling virus infections (38). Therefore, we addressed the question of whether these cells behave as CTLs ex vivo. Purified CD8+ cells were sorted into the distinct subsets and their cytotoxic capacity was tested in a CD3 mAb–mediated redirected cytotoxicity assay (Fig. 5). As expected, unprimed CD45RA+CD27+ cells were not able to lyse the target cells. CD45RACD27+ cells had a low but detectable cytolytic activity at relatively high effector/target ratios. Strikingly, however, the CD45RA+CD27 population had potent cytolytic activity.

Fas–Fas ligand interaction is an important mechanism used by CTLs for cell-mediated cytotoxicity (39, 40). Next to this, CTL effectors can kill target cells by releasing a pore-forming protein (perforin) and serine proteases (granzymes), which are stored in intracellular granules, into the vicinity of target cell membranes (41, 42). We analyzed freshly isolated CD8+ cells for the presence of perforin and granzyme B. In accordance with the cytolytic capacities of the different subsets, nearly all cells of the CD45RA+CD27 population contained perforin and granzyme B. Unprimed CD45RA+CD27+ cells did not contain these enzymes, whereas low staining could be found within the CD45RA population (Fig. 6 and Table 4). As already seen in the analysis of cytokine production, cells with dull CD27 expression showed a similar granzyme B and perforin expression pattern to CD27 cells.

The CD45RACD27+ Subset Contains Antigen-specific Precursor CTL.

CTL effectors exert direct ex vivo cytolytic activity, whereas memory cells need restimulation with antigen to acquire cytotoxic function. Since CD45RA CD27+ cells, which resemble memory T cells with respect to membrane phenotype and cytokine secretion ability, showed no direct ex vivo cytolytic activity, we studied whether cytotoxic effectors could be generated from this population. To this purpose, PBMCs from an HIV-1 seropositive individual were sorted into CD8+CD45RA+CD27+ and CD8+ CD45RACD27+ subsets. CTLs from this individual had been previously found to recognize an HLA-B57 restricted epitope of the HIV-1LAI RT aa 244-252 (Klein, M.R., unpublished results). Neither CD45RA+CD27+ nor CD45RA CD27+ cells showed direct ex vivo cytotoxicity as measured in a redirected kill assay as described above. To determine the frequency of peptide-specific CTLp within the distinct subsets, sorted cells were seeded in serial dilutions and stimulated with autologous B-LCL infected with recombinant vaccinia virus expressing the HIV-1LAI RT gene. On day 15, effector cells were tested for cytotoxicity on autologous B-LCL pulsed with the HIV-1LAI reverse transcriptase–derived peptide aa 244-252 (Fig. 7). Indeed, cytotoxic effector cells could be generated from the CD45RA CD27+ subset, which had an enhanced frequency of peptide-specific CTLps (61/106 cells) as compared to the CD45RA+CD27+ subset (15/106 cells).

CD45RACD27+ but not CD45RA+CD27 Support Ig Secretion In Vitro.

Apart from their cytolytic capacities, CD8+ cells have been shown to exert suppressor activity as well as helper activity for Ig synthesis (43). Helper capacity of CD8+ T cell subsets was tested in a CD3 mAb–driven T cell–dependent B cell differentiation assay (44). When total CD4+ and CD8+ cells were compared, helper activity was preferentially found in the CD4+ fraction (Fig. 8 A). However, when CD8+ cells were further separated into the indicated subsets, it was found that the CD45RACD27+ cells were also able to support B cell differentiation, whereas both the CD45RA+CD27+ and the CD45RA+CD27 cells showed no helper activity.

To test suppressor capacity, sorted CD8+ subsets were added to the autologous coculture of CD4+ and B cells (Fig. 8 B). In accordance with a previous study (43), total CD8+ T cells markedly reduced IgM and IgG synthesis in this system. When the CD8+ cells were further separated into the different subsets, suppressor activity was primarily found in the CD45RA+CD27+ fraction. This effect was not due to enhanced IL-4 consumption of CD8+CD45RA+ CD27+ cells, as addition of exogenous IL-4 did not overcome suppression of the Ig synthesis.

In summary, within the CD8+ compartment, helper and suppressor activity could be ascribed to CD45RACD27+ and CD45RA+CD27+ T cells, respectively, whereas the CD45RA+CD27 does not appear to directly influence B cell differentiation.

In this study, we define discrete subsets (summarized in Table 5) within the CD8+ T cell population based on expression patterns of CD45RA and CD27. First, similar to the CD4+ T cell subpopulation, CD45RA+CD27+ T cells are the only CD8+ lymphocytes present in cord blood, and the analysis of phenotype and function showed that this population primarily contains unprimed (naive) cells. CD8+CD45RACD27+ lymphocytes are T cells that have several features of antigen-experienced cells. In comparison to phenotypic data obtained in the murine system, these CD8+ cells most closely resemble memory-type cells, as they express intermediate levels of CD11a/CD18 and CD49d, but have no membrane expression of CD11b (9). Indeed, this CD8+CD45RACD27+ population from an HIV-1–infected individual contained, compared to CD8+ CD45RA+CD27+ T cells, an enhanced frequency of CTLp specific for an HIV-1 RT-derived peptide. In line with this, a study by Merkenschlager and Beverley (17) documented an elevated precursor frequency to recall antigens within the CD8+CD45RACD45R0+ fraction. A second population that has features of past antigen stimulation has a CD8+CD45RA+CD27 phenotype. Not only with respect to cytolytic potential, but also regarding surface expression of high levels of CD11b and CD49d and absence of the lymph node homing receptor CD62L, these cells fulfill the criteria of CTL effector-type cells that have been defined in murine models. Further characterization of the CD8+CD45RA+CD27 population allowed us to determine the functional abilities of human CTL-type effector cells as they occur in vivo. The data show that (a) on a per cell basis the vast majority of effector cells are able to produce both IFN-γ and TNF-α, (b) these cells contain high amounts of perforin and granzyme B, (c) Fas ligand mRNA is abundantly expressed in freshly isolated effector cells, and (d), that these cells exert potent cytotoxic activity without previous in vitro stimulation. Finally, a fourth population constituted of CD45RACD27 T cells can be found in the CD8+ fraction of all donors. The frequency of these cells in healthy donors is very low (usually <4%) and therefore not accessible for assays that depend on vast quantities of purified cells. However, based on membrane phenotype, cytokine production pattern, and expression of perforin and granzyme B, it is suggested that this population may contain both memory- and effector-type cells.

Based on the finding that the majority of CD8+CD45RA+ CD27 T cells lack CD28 expression, it was not surprising to find that this subset shares a number of features that have been previously reported for CD28 lymphocytes (15, 45, 46). Azuma et al. (15) showed that CD8+CD28 T cells are able to lyse FcR-bearing targets cells in the presence of CD3 mAb. Moreover, Berthou et al. recently reported that most of the perforin containing CD8+ lymphocytes lack the CD28 molecule (47). The current data confirm and extend these observations by showing that not only perforin but also granzyme B and Fas ligand are expressed within this T cell subset, which is likely to contribute to the lysis of target cells (39, 40). It is of interest to note that two major costimulatory receptors for T cell growth and cytokine production, CD28 and CD27, are absent from this effector-type T cell. The absence of CD28 implies that this cell type is unable to interact productively (in terms of induction of clonal expansion) with professional antigen-presenting cells. This inability is underscored by the observation that, in marked contrast to both the CD45RA+CD27+ and CD45RACD27+ subsets, CD45RA+CD27 T cells do not proliferate in response to IL-12, one of the major T cell stimulatory cytokines secreted by activated antigen-presenting cells (48). Finally, absence of CD27 on these cells also precludes costimulatory interactions with other, activated lymphocytes (49). Analysis of the proliferation requirements showed that CD45RA+CD27 T cells responded poorly to most stimuli, which is in agreement with functional studies on the CD8+CD28 T cell subset (15, 46). However, CD45RA+CD27 T cells do respond to IL-2 and to a lesser extent to IL-15, which suggests that in vivo this subset might be influenced by Th-derived signals.

In mice, the expression of CD49d nicely separates naive (low), memory (intermediate), and effector (high expression) T cells, and a study by Christensen et al. has shown that CD49d plays a critical role in efficient homing of effector T cells during virus infections (13). Also, in humans CD49d expression correlates well with the distinct CD8+ subsets, CD45RA+CD27 T cells being the ones with the highest expression. Apart from CD49d, a β1 integrin interacting with vascular cellular adhesion molecule–1 on activated endothelium and also the high expression of the β2 integrins CD11a and CD11b (Table 1) will probably influence the homing properties of these effector-type cells, which may be different from those of naive or memory cells. Indeed, CD8+CD45RA+CD27 cells were conspicuously absent from tonsils, whereas CD45RACD27+ cells could readily be demonstrated in this lymphoid tissue (Hamann, D., unpublished data). In line with this observation, it was reported that CD11b+ cells are present in blood, liver, and spleen, but absent from tonsil, lymph node, and thymus (45).

Thus, it would seem that in response to antigen, two types of primed CD8+ populations may develop. First, an effector-type population with high levels of granzyme, perforin, and Fas ligand expression that exerts cytolytic activity, which has lost the ability for autocrine proliferation but has retained the ability to synthesize IFN-γ and TNF-α, two cytokines implicated in the neutralization of viruses. Second, a population of memory-type cells, which is not cytolytic without further activation, produces a wide range of cytokines and is able to provide helper activity for B cell differentiation. Kinetic studies in mice showed that effector T cells can be found relatively early after virus infection (8 d), and decline thereafter. In contrast, memory cells become prominent at a later point (>50 d), and it has been suggested that effector cells seed the memory pool (9, 50). However, the relation between these subpopulations in humans is unclear at this moment. In major contrast to data obtained in the mouse system, effector-type T cell numbers in humans are relatively stable over time (>18 months, Rep, M.H.G., unpublished data). In addition, it has been reported that CD8+CD45RA+CD11abright, CD11b+, and CD28 T cells increase with age (45, 51, 52) and, in agreement with this (see Table 5 for phenotypic similarities), we found a similar relationship between age and the percentage of CD8+CD45RA+CD27 T cells (Rep, M.H.G., unpublished data). The reason for the discrepancy in the kinetics of effector-type T cells between mouse and human is not clear at this moment but it is possible that persistence of antigen, cross-reactive epitopes (53), and/or stimulatory cytokines plays a role (54). It can be imagined that these factors will more readily influence the maintenance of a polyclonal effector T cell population in a natural environment than that of the monoclonal T cell population present in laboratory animals housed under pathogen-free conditions. It is questionable, based on the in vitro experiments, whether effector T cells that are present in the human circulation have a sizable clonogenic potential. Still, it can be envisaged that this type of cell contributes to the inhibition of pathogen spread early in secondary infection through immediate lysis of infected cells.

In agreement with recent data on CD28 subsets by Monteiro et al. (55), we found shortened telomers in CD8+ CD45RA+CD27 compared to CD8+CD45RA+CD27+ in some donors (Hamann, D., K.C. Wolthers, S.A. Otto, P.A. Baars, F.M. Miedema, and R.A.W. van Lier, manuscript submitted for publication). This finding shows that transition to the CD45RA+CD27 subset is accompanied by cellular division. Cell cycle analysis showed that, analogous to the CD4+ population (23, 56), the CD8+ cells that are in G2M can only be found in the minute fraction of CD8+CD45RAbrightCD45R0bright cells and not in CD45RA+CD45R0CD27+ T cells (Baars, P.A., unpublished data). This finding implicates that CD45RA+CD27 cells could only develop via a CD45RA+CD45R0+ and a subsequent CD45RACD45R0+ stage. However, at this moment we cannot exclude that the transition from CD27+ to CD27 takes place in compartments, such as solid tissue, which are not readily available for cell-biological analysis. Finally, similar to what has been described for CD28 (57) and CD57+ (16, 37) T cells, we found a limited usage of Vβ elements by CD8+CD45RA+CD27 cells (Hamann, D., K.C. Wolthers, S.A. Otto, P.A. Baars, F.M. Miedema, and R.A.W. van Lier, manuscript submitted for publication). These findings, combined with the data on telomeric repeat length, infer that these circulating effector-type cells in humans are generated through antigen-dependent differentiation and proliferation processes.

Analysis of naive, memory, and CTL effector-type CD8+ T cells seems relevant not only in patients with chronic viral infections such as CMV, hepatitis B, or HIV-1, but also in patients actively immunized with tumor antigens and after having received an allotransplantation. Patients infected with CMV have been shown to have an increased percentage of CD57+ cells within the CD8+ population that contains virus-specific CTLs and can be oligoclonal (16). In HIV-1 infected individuals, an expanded CD28 population within the CD8+ T cell subset has been reported (46). This population has been found to be primarily CD57+, exerts potent cytolytic activity, and expresses perforin (46, 58). Interestingly, in contrast to healthy donors where CD27 and CD28 cells form highly overlapping subsets, a large population of CD45RACD28 cells that still express the CD27 antigen emerges during acute and persistent viral infections (Roos, M.T.L., manuscript submitted for publication), which may point to a sequential downregulation of costimulatory molecules during postthymic T cell differentiation.

Based on the data presented in this paper, we conclude that unidimensional analysis of CD8+ T cells, using either CD45RA, CD27, or CD28 mAbs, underestimates the complexity of this subset in humans. Therefore, we propose that combined staining with CD27 and CD45RA mAbs provides a tool not only for distinguishing unprimed CD45RA+CD27+ from primed CD45RA cells, but also for following the dynamics and expansion of a CD45RA+ CD27 cytotoxic effector cell population in CD8+ T cells in different disease states.

We would like to thank our colleagues Susanne Lens, Kiki Tesselaar, Wiebo van der Graaff, Marijke Roos, Peter Schellekens, and Frank Miedema for stimulating discussions and their help in preparing this manuscript.

1
Merkenschlager
M
,
Terry
L
,
Edwards
R
,
Beverley
PC
Limiting dilution analysis of proliferative responses in human lymphocyte populations defined by the monoclonal antibody UCHL1: implications for differential CD45 expression in T cell memory formation
Eur J Immunol
1988
18
1653
1661
[PubMed]
2
Byrne
JA
,
Butler
JL
,
Cooper
MD
Differential activation requirements for virgin and memory T cells
J Immunol
1988
141
3249
3257
[PubMed]
3
Sanders
ME
,
Makgoba
MW
,
June
CH
,
Young
HA
,
Shaw
S
Enhanced responsiveness of human memory T cells to CD2 and CD3 receptor-mediated activation
Eur J Immunol
1989
19
803
808
[PubMed]
4
Picker
LJ
,
Singh
MK
,
Zdraveski
Z
,
Treer
JR
,
Waldrop
SL
,
Bergstresser
PR
,
Maino
VC
Direct demonstration of cytokine synthesis heterogeneity among human memory/effector T cells by flow cytometry
Blood
1995
86
1408
1419
[PubMed]
5
Sanders
ME
,
Makgoba
MW
,
Sharrow
SO
,
Stephany
D
,
Springer
TA
,
Young
HA
,
Shaw
S
Human memory T lymphocytes express increased levels of three cell adhesion molecules (LFA-3, CD2, and LFA-1) and three other molecules (UCHL-1, CDw29, and Pgp-1) and have enhanced IFN-γ production
J Immunol
1988
140
1401
1407
[PubMed]
6
Akbar
AN
,
Terry
L
,
Timms
A
,
Beverley
PC
,
Janossy
G
Loss of CD45R and gain of UCHL1 reactivity is a feature of primed T cells
J Immunol
1988
140
2171
2178
[PubMed]
7
Michie
CA
,
McLean
A
,
Alcock
C
,
Beverley
PC
Life span of human lymphocyte subsets defined by CD45 isoforms
Nature (Lond)
1992
360
264
265
[PubMed]
8
Bell
EB
,
Sparshott
SM
Interconversion of CD45R subsets of CD4 T cells in vivo
Nature (Lond)
1990
348
163
166
[PubMed]
9
Zimmermann
C
,
Brduscha-Riem
K
,
Blaser
C
,
Zinkernagel
RM
,
Pircher
H
Visualization, characterization, and turnover of CD8+memory T cells in virus-infected hosts
J Exp Med
1996
183
1367
1375
[PubMed]
10
Mobley
JL
,
Dailey
MO
Regulation of adhesion molecule expression by CD8 T cells in vivo. I. Differential regulation of gp90MEL-14 (LECAM-1), Pgp-1, LFA-1, and VLA-4 α during the differentiation of cytotoxic T lymphocytes induced by allografts
J Immunol
1992
148
2348
2356
[PubMed]
11
McFarland
HI
,
Nahill
SR
,
Maciaszek
JW
,
Welsh
RM
CD11b (Mac-1): a marker for CD8+cytotoxic T cell activation and memory in virus infection
J Immunol
1992
149
1326
1333
[PubMed]
12
Andersson
EC
,
Christensen
JP
,
Marker
O
,
Thomsen
AR
Changes in cell adhesion molecule expression on T cells associated with systemic virus infection
J Immunol
1994
152
1237
1245
[PubMed]
13
Christensen
JP
,
Andersson
EC
,
Scheynius
A
,
Marker
O
,
Thomsen
AR
α4 integrin directs virus-activated CD8+T cells to sites of infection
J Immunol
1995
154
5293
5301
[PubMed]
14
Walker
PR
,
Ohteki
T
,
Lopez
JA
,
MacDonald
HR
,
Maryanski
JL
Distinct phenotypes of antigen-selected CD8 T cells emerge at different stages of an in vivo immune response
J Immunol
1995
155
3443
3452
[PubMed]
15
Azuma
M
,
Phillips
JH
,
Lanier
LL
CD28−T lymphocytes. Antigenic and functional properties
J Immunol
1993
150
1147
1159
[PubMed]
16
Wang
ECY
,
Moss
PA
,
Frodsham
P
,
Lehner
PJ
,
Bell
JI
,
Borysiewicz
LK
CD8highCD57+T lymphocytes in normal, healthy individuals are oligoclonal and respond to human cytomegalovirus
J Immunol
1995
155
5046
5056
[PubMed]
17
Merkenschlager
M
,
Beverley
PC
Evidence for differential expression of CD45 isoforms by precursor for memory-dependent and independent cytotoxic responses: human CD8 memory CTLp selectively express CD45R0 (UCHL1)
Int Immunol
1989
1
450
459
[PubMed]
18
De Jong
R
,
Brouwer
M
,
Miedema
F
,
van Lier
RAW
Human CD8+ T lymphocytes can be divided into CD45RA+ and CD45R0+cells with different requirements for activation and differentiation
J Immunol
1991
146
2088
2094
[PubMed]
19
Roederer
M
,
Dubs
JG
,
Anderson
MT
,
Raju
PA
,
Herzenberg
LA
CD8 naive T cell counts decrease progressively in HIV-infected adults
J Clin Invest
1995
95
2061
2066
[PubMed]
20
Okumura
M
,
Fujii
Y
,
Inada
K
,
Nakahara
K
,
Matsuda
H
Both CD45RA+ and CD45RA− subpopulations of CD8+T cells contain cells with high levels of lymphocyte function-associated antigen-1 expression, a phenotype of primed T cells
J Immunol
1993
150
429
437
[PubMed]
21
Hintzen
RQ
,
De Jong
R
,
Lens
SMA
,
Brouwer
M
,
Baars
P
,
van Lier
RAW
Regulation of CD27 expression on subsets of mature T-lymphocytes
J Immunol
1993
151
2426
2435
[PubMed]
22
De Jong
R
,
Brouwer
M
,
Hooibrink
B
,
van der Pouw-Kraan
T
,
Miedema
F
,
van Lier
RAW
The CD27− subset of peripheral blood memory CD4+lymphocytes contains functionally differentiated T lymphocytes that develop by persistent antigenic stimulation in vivo
Eur J Immunol
1992
22
993
999
[PubMed]
23
Baars
PA
,
Maurice
MM
,
Rep
M
,
Hooibrink
B
,
van Lier
RAW
Heterogeneity of the circulating human CD4+ T cell population: further evidence that the CD4+ CD45RA−CD27−T cell subset contains specialized primed T cells
J Immunol
1995
154
17
25
[PubMed]
24
van der Pouw-Kraan
T
,
van Kooten
C
,
Rensink
I
,
Aarden
L
IL-4 production by human T cells. Differential regulation of IL-4 versus IL-2 production
Eur J Immunol
1992
22
1237
1241
[PubMed]
25
Takahashi
T
,
Tanaka
M
,
Inazawa
J
,
Abe
T
,
Suda
T
,
Nagata
S
Human Fas ligand: gene structure, chromosomal localization and species specificity
Int Immunol
1994
6
1567
1572
[PubMed]
26
Jolly
DJ
,
Okayama
H
,
Berg
P
,
Esty
AC
,
Filpula
D
,
Bohlen
P
,
Johson
GG
,
Shivelly
JE
,
Hunkapillar
T
,
Friedman
T
Isolation and characterization of a full-length expressible cDNA for human hypoxanthine phosphoribosyltransferase
Proc Natl Acad Sci USA
1983
80
477
482
[PubMed]
27
Jung
T
,
Schauer
U
,
Heusser
C
,
Neuman
C
,
Rieger
C
Detection of intracellular cytokines by flow cytometry
J Immunol Methods
1993
159
197
207
[PubMed]
28
Hamann
D
,
Hilkens
CMU
,
Grogan
JL
,
Lens
SMA
,
Kapsenberg
ML
,
Yazdanbakhsh
M
,
van Lier
RAW
CD30 expression does not discriminate between human Th- and Th-2 type T cells
J Immunol
1996
156
1387
1391
[PubMed]
29
Gillis
S
,
Ferm
MM
,
Ou
W
,
Smith
KA
T-cell growth factor: parameters of production and a quantitative microassay for activity
J Immunol
1978
120
2027
2033
[PubMed]
30
De Jong
R
,
Brouwer
M
,
Rebel
VI
,
Van Seventer
GA
,
Miedema
F
,
van Lier
RAW
Generation of alloreactive cytolytic T lymphocytes by immobilized anti-CD3 monoclonal antibodies. Analysis of requirements for human cytolytic T-lymphocyte differentiation
Immunology
1990
70
357
364
[PubMed]
31
van Baalen
CA
,
Klein
MR
,
Geretti
AM
,
Keet
RIPM
,
Miedema
F
,
Van Els
CACM
,
Osterhaus
ADME
Selective in vitro expansion of HLA class I–restricted HIV-1 Gag-specific CD8+T cells: cytotoxic T-lymphocyte epitopes and precursor frequencies
AIDS (Phila)
1993
7
781
786
[PubMed]
32
Klein
MR
,
Holwerda
AM
,
van Baalen
CA
,
Bende
RJ
,
Kerkhof
SR
,
Osterhaus
ADME
,
Keet
IPM
,
Eeftinck
JKM
,
Scattenkerk
,
Schuitemaker
H
,
Miedema
F
Kinetics of Gag-specific cytotoxic T lymphocyte responses during the clinical course of HIV-1 infection: a longitudinal analysis of rapid progressors and long-term asymptomatics
J Exp Med
1995
181
1365
1372
[PubMed]
33
Strijbosch
LWG
,
Does
RJMM
,
Buurman
WA
Computer-aided design and evaluation of limiting and serial dilution experiments
Int J Bio-Med Comput
1988
23
279
290
[PubMed]
34
Hamann
D
,
Baars
PA
,
Hooibrink
B
,
van Lier
RAW
Heterogeneity of the human CD4+ T-cell population: two distinct CD4+T-cell subsets characterized by coexpression of CD45RA and CD45R0 isoforms
Blood
1996
9
3513
3521
[PubMed]
35
Salmon
M
,
Kitas
GD
,
Bacon
PA
Production of lymphokine mRNA by CD45R+ and CD45R− helper T cells from human peripheral blood and by human CD4+T cell clones
J Immunol
1989
143
907
912
[PubMed]
36
Adamthwaite
D
,
Cooley
MA
CD8+T-cell subsets defined by expression of CD45 isoforms differ in their capacity to produce Il-2, IFN-γ and TNF-β
Immunology
1994
81
253
260
[PubMed]
37
Wang
ECY
,
Taylor-Wiedeman
J
,
Perera
P
,
Fisher
J
,
Borysiewicz
LK
Subsets of CD8+, CD57+cells in normal, healthy individuals: correlation with human cytomegalovirus (HCMV) carrier status, phenotypic and functional analyses
Clin Exp Immunol
1993
94
297
305
[PubMed]
38
Guidotti
LG
,
Ishikawa
T
,
Hobbs
MV
,
Matzke
B
,
Schreiber
R
,
Chisari
FV
Intracellular inactivation of the hepatitis B virus by cytotoxic T lymphocytes
Immunity
1996
4
25
36
[PubMed]
39
Kägi
D
,
Vignaux
F
,
Lederman
B
,
Burki
K
,
Depraetere
V
,
Nagata
S
,
Hengartner
H
,
Golstein
P
Fas and perforin pathways as major mechanisms of T-cell mediated cytotoxicity
Science (Wash DC)
1994
265
528
530
[PubMed]
40
Lowin
B
,
Hahne
M
,
Mattman
C
,
Tshopp
J
Cytolytic T-cell cytotoxicity is mediated through perforin and Fas lytic pathways
Nature (Lond)
1994
370
650
652
[PubMed]
41
Smyth
MJ
,
Trapani
JA
Granzymes: exogenous proteinases that induce target cell apoptosis
Immunol Today
1995
16
202
206
[PubMed]
42
Liu
CC
,
Walsh
CM
,
Young
JD
Perforin: structure and function
Immunol Today
1995
16
194
201
[PubMed]
43
Rieber
EP
,
Rank
G
CDw60: a marker for human CD8+T helper cells
J Exp Med
1994
179
1385
1390
[PubMed]
44
van Lier
RAW
,
Brouwer
M
,
Rebel
VI
,
van Noesel
CJ
,
Aarden
LA
Immobilized anti-CD3 monoclonal antibodies induce accessory cell–independent lymphokine production, proliferation and helper activity in human T lymphocytes
Immunology
1989
68
45
50
[PubMed]
45
Hoshino
T
,
Yamada
A
,
Honda
J
,
Imai
Y
,
Nakao
M
,
Inoue
M
,
Sagawa
K
,
Yokoyama
MM
,
Oizumi
K
,
Itoh
K
Tissue-specific distribution and age-dependent increase of human CD11b+T cells
J Immunol
1993
151
2237
2246
[PubMed]
46
Borthwick
NJ
,
Bofill
M
,
Gombert
WM
,
Akbar
AN
,
Medina
E
,
Sagawa
K
,
Lipman
MC
,
Johnson
MA
,
Janossy
G
Lymphocyte activation in HIV-1 infection. II. Functional defects of CD28−T cells
AIDS (Phila)
1994
8
431
441
[PubMed]
47
Berthou
C
,
Legros-Maida
S
,
Soulie
A
,
Wargnier
A
,
Guillet
J
,
Rabian
C
,
Gluckman
E
,
Sasportes
M
Cord blood T lymphocytes lack constitutive perforin expression in contrast to adult peripheral blood T lymphocytes
Blood
1995
85
1540
1546
[PubMed]
48
Trinchieri
G
Interleukin-12: a cytokine produced by antigen-presenting cells with immunoregulatory functions in the generation of T-helper cells type 1 and cytotoxic lymphocytes
Blood
1994
84
4008
4027
[PubMed]
49
Hintzen
RQ
,
De Jong
R
,
Lens
SMA
,
van Lier
RAW
CD27: marker and mediator of T-cell activation
Immunol Today
1994
15
307
310
[PubMed]
50
Bruno
L
,
Kirberg
J
,
Von Boehmer
H
On the cellular basis of immunological T cell memory
Immunity
1995
2
37
43
[PubMed]
51
Okumura
M
,
Fujii
Y
,
Takeuchi
Y
,
Inada
K
,
Nakahara
K
,
Matsuda
H
Age-related accumulation of LFA-1high cells in a CD8+CD45RAhighT cell population
Eur J Immunol
1993
23
1057
1063
[PubMed]
52
Fagnoni
FF
,
Vescovini
R
,
Mazzola
M
,
Bologna
G
,
Nigro
E
,
Lavagetto
G
,
Franceshi
C
,
Passeri
M
,
Sansoni
P
Expansion of CD8+CD28−T cells in healthy ageing people, including centenarians
Immunology
1996
88
501
507
[PubMed]
53
Beverley
PC
Is T-cell memory maintained by crossreactive stimulation?
Immunol Today
1990
11
203
205
[PubMed]
54
Tough
DF
,
Borrow
P
,
Sprent
J
Induction of bystander T cell proliferation by viruses and type I interferon in vivo [see comments]
Science (Wash DC)
1996
272
1947
1950
[PubMed]
55
Monteiro
J
,
Batliwalla
F
,
Ostrer
H
,
Gregersen
PK
Shortened telomeres in clonally expanded CD28− CD8+ T cells imply a replicative history that is distinct from their CD28+CD8+counterparts
J Immunol
1996
156
3587
3590
[PubMed]
56
Picker
LJ
,
Treer
JR
,
Ferguson-Darnell
B
,
Collins
PA
,
Buck
D
,
Terstappen
LWMM
Control of lymphocyte regulation in man. I. Differential regulation of the peripheral lymph node homing receptor L-selectin on T cells during the virgin to memory transition
J Immunol
1993
150
1105
1121
[PubMed]
57
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
[PubMed]
58
Vingerhoets
JH
,
Vanham
GL
,
Kestens
LL
,
Penne
GG
,
Colebunders
RL
,
Vandenbruaene
MJ
,
Goeman
J
,
Gigase
PL
,
De Boer
M
,
Ceuppens
JL
Increased cytolytic T lymphocyte activity and decreased B7 responsiveness are associated with CD28 down-regulation on CD8+T cells from HIV-infected subjects
Clin Exp Immunol
1995
100
425
433
[PubMed]

D. Hamann was supported by the Human Capital and Mobility Programme of the European Community, and P.A. Baars and M.H.G. Rep were supported by the Dutch Society for Support of Research on Multiple Sclerosis.

1

Abbreviations used in this paper: B-LCL, B lymphoblastoid cell lines; p, precursors; RT, reverse transcriptase.

Author notes

Address correspondence to René A.W. van Lier, Department of Clinical Viro-Immunology, Central Laboratory of the Netherlands Red Cross Blood Transfusion Service, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands. Phone: 31-20-5123317; FAX: 31-20-5123310.