Whereas most peripheral CD8+ αβ T cells highly express CD8αβ heterodimer in healthy individuals, there is an increase of CD8α+βlow or CD8αα αβ T cells in HIV infection or Wiskott-Aldrich syndrome and after bone marrow transplantation. The significance of these uncommon cell populations is not well understood. There has been some question as to whether these subsets and CD8α+βhigh cells belong to different ontogenic lineages or whether a fraction of CD8α+βhigh cells have down-regulated CD8β chain. Here we assessed clonality of CD8αα and CD8α+βlow αβ T cells as well as their phenotypic and functional characteristics. Deduced from surface antigens, cytotoxic granule constituents, and cytokine production, CD8α+βlow cells are exclusively composed of effector memory cells. CD8αα cells comprise effector memory cells and terminally differentiated CD45ROCCR7memory cells. T-cell receptor (TCR) Vβ complementarity-determining region 3 (CDR3) spectratyping analysis and subsequent sequencing of CDR3 cDNA clones revealed polyclonality of CD8α+βhigh cells and oligoclonality of CD8α+βlow and CD8αα cells. Importantly, some expanded clones within CD8αα cells were also identified within CD8α+βhigh and CD8α+βlow subpopulations. Furthermore, signal-joint TCR rearrangement excision circles concentration was reduced with the loss of CD8β expression. These results indicated that some specific CD8α+βhigh αβ T cells expand clonally, differentiate, and simultaneously down-regulate CD8β chain possibly by an antigen-driven mechanism. Provided that antigenic stimulation directly influences the emergence of CD8αα αβ T cells, these cells, which have been previously regarded as of extrathymic origin, may present new insights into the mechanisms of autoimmune diseases and immunodeficiencies, and also serve as a useful biomarker to evaluate the disease activities.

CD8 is a coreceptor that recognizes the nonpolymorphic α3 domain of the major histocompatibility complex (MHC) class I molecules and is necessary for T-cell activation.1,2 It increases the avidity of the interaction between the CD8-bearing T cell and the antigen-presenting cell.1,3,4 With the T-cell receptor (TCR)–peptide-MHC ligation, simultaneous coligation of the coreceptor juxtaposes MHC-engaged TCR complexes with intracellular signaling intermediates, leading to increased tyrosine phosphorylation and further recruitment and activation of downstream signaling effector molecules.2,5-7 CD8 antigen is composed of 2 kinds of molecules, α and β chain, and is expressed either as an αα homodimer or an αβ heterodimer.8-11 These isoforms are the products of closely linked but distinct genes exhibiting only moderate sequence homology.12,13 Studies of CD8α and CD8β have revealed the distinct contributions to the coreceptor function. CD8α can interact with all molecules presently known to be involved in CD8 function by itself. CD8β, on the contrary, has roles to make the coreceptor function more efficiently as CD8αβ heterodimers. Extracellular domain of CD8β increases the avidity of CD8 binding to MHC class I14 and influences specificity of the CD8/MHC/TCR interaction.15 CD8β may also uniquely mediate efficient interaction with the TCR/CD3 complex.16In addition, the intracellular domain of CD8β enhances association of CD8α with Lck and linker for activation of T cells (LAT).14,17,18 

In healthy individuals, most thymocytes and peripheral T cells highly express the heterodimeric form of CD8.17 These CD8α+βhigh T cells express not only CD8αβ heterodimers but also CD8αα homodimers on the same cells.9,10 On the other hand, specific subpopulations of natural killer (NK) cells and intestinal γδ T cells exclusively express CD8αα.17 However, CD8α+βlow and CD8αα αβ T cells increase in the periphery in some conditions. Patients with Wiskott-Aldrich syndrome (WAS) are reported to have CD8+ T cells composed mostly of CD8αα homodimers.19 Also, a large proportion of CD8+ T cells reconstituted in bone marrow transplant recipients express CD8αα homodimers.20,21 In addition, HIV infection is characterized by the appearance of a major CD8 subpopulation with reduced CD8β chains, which exhibits strong antiviral activity.22 

Although there has been much controversy as to the origin and the functional roles of these cells, there is increasing evidence in recent literature to suggest that CD8αα αβ T cells derive from the thymus after positive selection and that they exhibit distinct functions from conventional CD8αβ αβ T cells.23,24Furthermore, it seems that expression of CD8α chains is secondarily regulated by the intestinal microenvironments.25 However, despite the extensive studies of CD8α and β chains in vitro and the studies on a molecular basis, heterogeneity of CD8 isoform expression may not have been examined thoroughly in various human disorders and clinical conditions. Moreover, the in vivo function and clinical significance of CD8αα and CD8α+βlowαβ T cells are poorly understood. The purpose of this study is to reveal in vivo cell function and the origin of CD8αα and CD8α+βlow αβ T cells. More specifically, we analyzed cell surface antigen expression, cytotoxic granule constituents, and cytokine production of these subpopulations. Furthermore, this study also examined if CD8αα and CD8α+βlow αβ T cells comprise distinct clones, or if they descend directly from CD8α+βhigh cells by down-regulating CD8β chain after antigen stimulation in vivo.

Monoclonal antibodies

Fluorescein isothiocyanate (FITC)–conjugated monoclonal antibodies (mAbs) recognizing CD95, CD45RO, and R-phycoerythrin-Cyanine5 (RPE-Cy5)–conjugated anti-CD8α mAb were purchased from Dako (Glostrup, Denmark). FITC-conjugated mAbs against CD16, CD27, CD57, TCRαβ, interleukin 2 (IL-2), interferon γ (IFN-γ), and mouse IgG antibodies as well as nonconjugated anti-CCR7 mAbs were obtained from BD Pharmingen (San Diego, CA). FITC-conjugated anti-CD28, anti-TCRγδ, anti-CD62L, PE-conjugated anti-CD8β, anti-2B4, anti–TIA-1 (a cytotoxic granule-associated protein), and nonconjugated anti-CD8β mAbs were products of Beckman Coulter (Tokyo, Japan). PE-conjugated mAbs against perforin and granzyme B were purchased from Ancell (Bayport, MN) and Research Diagnostics (Flanders, NJ), respectively.

Cell preparation and flow cytometric analysis

Human peripheral blood mononuclear cells (PBMNCs) were isolated from heparinized peripheral blood by Ficoll-Hypaque density centrifugation. CD16+ and TCRγδ+ cells were then depleted using MACS and anti-FITC magnetic beads (Miltenyi Biotec, Bergisch Gladbach, Germany) after staining with FITC-conjugated anti-TCRγδ and anti-CD16 mAbs. The negatively sorted cells (purity > 99%) were stained with PE-conjugated anti-CD8β and RPE-Cy5–conjugated anti-CD8α mAb in combination with FITC-conjugated anti-TCRαβ, anti-CD62L, anti-CD57, anti-CD95, anti–HLA-DR, or anti-CD45RO mAbs. For the analysis of CCR7 expression, nonconjugated anti-CCR7 mAbs were used with FITC-conjugated goat antimouse antibodies. Similarly, 2B4 expression was analyzed using FITC-conjugated goat antimouse antibodies with the staining with nonconjugated anti-CD8β mAbs and PE-conjugated anti-2B4 mAbs. These stained cells, after washing with phosphate-buffered saline (PBS), were analyzed on a FACSCalibur flow cytometer (BD Biosciences, Tokyo, Japan). In addition, for signal-joint TCR rearrangement excision circles (Sj TRECs) quantification and TCR complementarity-determining region 3 (CDR3) spectratyping and sequencing, CD8α+αβ T cells with different (high, low, or negative) CD8β expression were separated using an Epics ELITE flow cytometer (Coulter Electronics, Hialeah, FL) after depletion of CD4+, CD14+, CD16+, CD20+, and TCRγδ+ cells with MACS (purity > 98%). Patterns of flow cytometric analysis pursued for 3 to 6 independent donors were similarly otherwise noted, and the representative results were presented.

Flow cytometric detection of cytokine production and intracellular staining for cytotoxic granule constituents

TCRγδ-depleted and CD16-depleted PBMNCs (TCRγδ CD16 PBMNCs) were stimulated for 6 hours with 10 ng/mL phorbol myristate acetate (PMA) and 500 ng/mL A23187 in the presence of 1 μg/mL monensin (Sigma, St Louis, MO). After cell surface staining with PE-conjugated CD8β and RPE-Cy5–conjugated CD8α, cells were fixed and permeabilized with Cytofix/Cytoperm Plus Kit (BD Pharmingen) per the manufacturer's instruction. Staining of the cytoplasm with FITC-conjugated anti–IFN-γ or anti–IL-2 mAb followed. Separately, freshly isolated TCRγδCD16 PBMNCs were treated with anti-CD8β mAb followed by FITC-conjugated goat antimouse antibodies. They were further stained with RPE-Cy-5–conjugated anti-CD8α mAbs after blocking with normal mouse serum. After fixation and permeabilization, the cells were stained with PE-conjugated antiperforin, antigranzyme B, or anti–TIA-1 mAbs.

RNA extraction and cDNA preparation

Total RNA was extracted from separated CD8+ αβ T cells with TRIZOL reagent following the manufacturer's instructions (Gibco BRL, Bethesda, MD). The RNA was then reverse-transcribed into cDNA in a reaction primed with oligo(dt)12-18 using SuperScript II reverse transcriptase as recommended by the manufacturer (Gibco BRL).

Sj TREC quantification

Sj TRECs were quantified in sorted CD8+ αβ T-cell subsets by a real-time quantitative polymerase chain reaction (PCR) method as described previously.26,27 Sorted cells were lysed in 100 μg/mL proteinase K (Wako Pure Chemical Industries, Osaka, Japan) for 1 hour at 56°C and then 10 minutes at 95°C at 107 cells/mL. Then PCR was carried out on 5 μL cell lysate in a spectrofluorometric thermal cycler (ABI PRISM 7700, Applied Biosystems, Osaka, Japan) under the following conditions: 50°C for 2 minutes followed by 95°C for 10 minutes, after which 50 cycles of amplification were carried out (95°C for 15 seconds, 60°C for 1 minute). The sequences of the primers and probe used were the following: forward primer GGAAAACACAGTGTGACATGGA, reverse primer GTCAACAAAGGTGATGCCACAT, and the probe FAM-CCTGTCTGCTCTTCATTCACCGTTCTCA-TAMRA. A standard curve was plotted, and Sj TREC values for samples were calculated by ABI PRISM 7700 software.

CDR3 spectratyping

CDR3 spectratyping was pursued as previously described.28 Briefly, cDNA was amplified by PCR through 35 cycles (94°C for 1 minute, 55°C for 1 minute, and 72°C for 1 minute) with a primer specific to 24 different BV subfamilies (BVs 1-2029 and BVs21-2430) and a fluorescent BC primer.29 The fluorescent PCR products were mixed with formamide and the size standard (GeneScan-500 TAMRA, Applied Biosystems). After denaturation for 2 minutes at 90°C, the products were analyzed with an automated 310 DNA sequencer (Applied Biosystems), and the data were analyzed with GeneScan software (Applied Biosystems).

The overall complexity within a Vβ subfamily was determined by counting the numbers of discrete peaks and determining their relative size on the spectratype histogram. We used a complexity scoring system31 with our interpretation, that is, complexity score = (sum of all the peak heights/sum of the major peak heights) × (number of the major peaks). Major peaks were defined as those peaks on the spectratype histogram whose amplitude was at least 10% of the sum of all the peak heights.

Cloning and sequencing of PCR-amplified cDNA

The PCR products of some BV cDNA were electrophoresed on an agarose gel and purified using QIAquick Gel Extraction Kit (Qiagen, Tokyo, Japan), and then cloned with TOPO TA Cloning (Invitrogen, Carlsbad, CA). Eleven to 19 colonies containing the insert fragment were randomly selected. Purified with QIAprep Spin Miniprep Kit (Qiagen), the recombinant plasmids were subjected to fluorescence dye terminator cycle sequencing, and the sequence reactions were analyzed on a 310 DNA sequencer (Applied Biosystems) after removal of the unincorporated fluorescence dye with Centri-Sep Spin Columns (Applied Biosystems).

Statistical analysis

Association of the percentage of peripheral CD8α+βlow and CD8αα αβ T cells with age was analyzed using the Spearman rank correlation coefficient. The Wilcoxon signed rank test was applied to examine statistically significant differences of CDR3 complexity scores between subpopulations of different CD8β expression.

CD8α+βlow and CD8αα αβ T cells expand with advancing age

To ensure that the number of peripheral CD8α+βlow and CD8αα αβ T cells are limited in healthy individuals, we first stained PBMNCs with anti-TCRαβ, anti-CD8β, and anti-CD8α mAbs conjugated to different fluorochromes in several healthy individuals including cord blood. CD8α+ TCRαβ+ cells could be classified into 3 groups defined by the level of CD8β expression: CD8α+βhigh, CD8 α+βlow, and CD8 α+β, which is CD8αα. Although CD8αα αβ T cells were negligible and small numbers of CD8α+βlow αβ T cells existed in cord blood, these populations increased in a 5-year-old child and even more in an adult (Figure 1). To assess the developmental changes of CD8α+βlow and CD8αα αβ T cells, we evaluated the frequency of these subpopulations in various age groups using more blood samples. In cord blood, CD8α+βlow and CD8αα αβ T cells represented a minor population within CD8α+αβ T cells. These subpopulations increased with advancing age as expected (P < .01). However, it is notable that some adults showed levels of CD8α+βlow and CD8αα αβ T cells as low as neonates (Figure2).

Fig. 1.

CD8β expression on CD8α+ αβ T cells in healthy individuals.

PBMNCs from healthy individuals and cord blood were stained with FITC-conjugated anti-TCRαβ, PE-conjugated anti-CD8β, and RPE-Cy5–conjugated anti-CD8α mAbs. TCRαβ and CD8α gated cells were analyzed for the expression of CD8α (y-axis) versus CD8β (x-axis). Representative data are displayed.

Fig. 1.

CD8β expression on CD8α+ αβ T cells in healthy individuals.

PBMNCs from healthy individuals and cord blood were stained with FITC-conjugated anti-TCRαβ, PE-conjugated anti-CD8β, and RPE-Cy5–conjugated anti-CD8α mAbs. TCRαβ and CD8α gated cells were analyzed for the expression of CD8α (y-axis) versus CD8β (x-axis). Representative data are displayed.

Close modal
Fig. 2.

Developmental change of CD8α+βlow and CD8αα fractions within CD8α+ αβ T cells.

CD8α+ αβ T cells were analyzed for CD8β expression, and the total frequencies of CD8α+βlow and CD8αα fractions were plotted along different age groups.

Fig. 2.

Developmental change of CD8α+βlow and CD8αα fractions within CD8α+ αβ T cells.

CD8α+ αβ T cells were analyzed for CD8β expression, and the total frequencies of CD8α+βlow and CD8αα fractions were plotted along different age groups.

Close modal

Correlation of CD8β expression with other surface markers

We next compared the expression of the various surface antigen markers on CD8α+ αβ T cells with different levels of CD8β expression. Before pursuing 3-color flow cytometric analysis, we depleted CD16+ NK cells and TCRγδ+ T cells from PBMNCs because these cells contain CD8α+ cells. The depletion of CD16+ and TCRγδ+ cells yielded TCRαβ+ or CD3+ cells with more than 98% purity when gated on CD8α (Figure 3A). CD8α+βhigh cells were heterogeneous for the expression of all the surface antigens analyzed. In the CD8α+βlow subpopulation, CD95+, CD45RO+, and 2B4+ cells became dominant, and the subset lost CD62L and CCR7 antigens. Most CD8αα T cells expressed CD95 and 2B4, but not CD57, CD62L, or CCR7. Although more than half of 7 adults analyzed had CD8αα cells, which exclusively expressed CD45RO, CD27, and CD28, the rest of the individuals possessed CD8αα cells that were as much as 30% negative for these surface antigens (Figure 3B and data not shown).

Fig. 3.

Analysis of surface antigen expression on CD8α+ αβ T cells.

TCRγδ CD16 PBMNCs were stained with CD8β, CD8α, and TCRαβ or CD3 (A), or other various surface antigens as indicated (B). CD8α gated cells are displayed.

Fig. 3.

Analysis of surface antigen expression on CD8α+ αβ T cells.

TCRγδ CD16 PBMNCs were stained with CD8β, CD8α, and TCRαβ or CD3 (A), or other various surface antigens as indicated (B). CD8α gated cells are displayed.

Close modal

Cytotoxic granule proteins and cytokine production

To further characterize the subpopulations of CD8+αβ T cells with regard to CD8β-chain expression, we analyzed CD8+ αβ T cells for the presence of perforin, granzyme B, and TIA-1. CD8α+βhigh cells were heterogeneous for the expression of the cytotoxic granule constituents. CD8α+βlow cells were also heterogeneous for the expression of perforin and granzyme B, but the subset entirely expressed TIA-1. A large number of CD8αα T cells possessed perforin and nearly all the cells contained TIA-1, whereas CD8αα cells did not contain granzyme B (Figure 4A).

Fig. 4.

Cytotoxic granule constituents and cytokine production.

(A) TCRγδ CD16 PBMNCs were stained with anti-CD8β mAbs recognized by FITC-conjugated goat-antimouse antibodies, RPE-Cy5–conjugated anti-CD8α, and PE-conjugated antiperforin, antigranzyme B, or anti–TIA-1 mAbs. (B) After the stimulation with PMA and A23187 in the presence of monensin, TCRγδCD16 PBMNCs were stained with PE-conjugated anti-CD8β, RPE-Cy5–conjugated anti-CD8α, and FITC-conjugated anti–IL-2 or anti–IFN-γ mAbs. CD8α gated cells are displayed.

Fig. 4.

Cytotoxic granule constituents and cytokine production.

(A) TCRγδ CD16 PBMNCs were stained with anti-CD8β mAbs recognized by FITC-conjugated goat-antimouse antibodies, RPE-Cy5–conjugated anti-CD8α, and PE-conjugated antiperforin, antigranzyme B, or anti–TIA-1 mAbs. (B) After the stimulation with PMA and A23187 in the presence of monensin, TCRγδCD16 PBMNCs were stained with PE-conjugated anti-CD8β, RPE-Cy5–conjugated anti-CD8α, and FITC-conjugated anti–IL-2 or anti–IFN-γ mAbs. CD8α gated cells are displayed.

Close modal

Because cytokine production capacity is also a major factor determining cell functions, CD8+ αβ T cells were stimulated for 6 hours with PMA and calcium ionophore in the presence of monensin for analysis of IFN-γ and IL-2 production. Heterogeneity for the cytokine production was observed in the CD8α+βhighsubset. Entire CD8α+βlow cells produced IFN-γ, and some proportion of the cells produced IL-2. CD8αα cells exclusively expressed IFN-γ, but not IL-2 (Figure 4B).

CD8α+βlow and CD8αα αβ T cells exhibit less clonal diversity

Sequence analysis of CDR3 length diversity in CD8α+βhigh, CD8α+βlow, and CD8αα αβ T cells was pursued to define the extent of clonal expansion. About 5 × 105 cells of each subpopulation were isolated, and their cDNA was subjected to PCR amplification with 24 Vβ-specific primers. TCR spectratypes of CD8α+βhighcells exhibited, with a few exceptions, a gaussianlike distribution, indicating that the subset comprises cells with highly diverse and polyclonal TCR repertoires. The profile of CD8α+βlow cells revealed skewed CDR3 size distribution in some Vβ subfamilies, but about one third of Vβ subfamilies remained diverse. To a further extent, the majority of Vβ subfamilies of CD8αα cells displayed apparently skewed patterns, many of them with an almost single peak pattern (Figure5).

Fig. 5.

Spectratypes of the T-cell repertoire within CD8α+βhigh, CD8α+βlow, and CD8αα αβ T cells.

Histograms of the relative sizes of the PCR-amplified CDR3 region within CD8α+βhigh, CD8α+βlow, and CD8αα αβ T cells in one donor are shown. The y-axis is relative quantity of RNA bearing the specific TCR Vβ. The x-axis represents the nucleotide length of the PCR-amplified TCR gene products.

Fig. 5.

Spectratypes of the T-cell repertoire within CD8α+βhigh, CD8α+βlow, and CD8αα αβ T cells.

Histograms of the relative sizes of the PCR-amplified CDR3 region within CD8α+βhigh, CD8α+βlow, and CD8αα αβ T cells in one donor are shown. The y-axis is relative quantity of RNA bearing the specific TCR Vβ. The x-axis represents the nucleotide length of the PCR-amplified TCR gene products.

Close modal

To quantify differences in the TCR Vβ gene repertoire among the T-cell subsets, we assigned complexity scores to each sample analyzed. Samples from 2 donors were presented; one of them (donor 1) did not possess CD8αα αβ T cells enough to be isolated. In donor 1, complexity scores of CD8α+βlowcells were significantly lower than CD8α+βhigh cells (P < .01). Likewise, complexity scores of CD8α+ αβ T cells in donor 2 decreased as they lost CD8β expression (CD8α+βhigh versus CD8α+βlow cells, P < .05; CD8α+βlow versus CD8αα cells,P < .001; Figure 6). These results suggest that CD8α+βlow and, to a larger extent, CD8αα αβ T cells comprise oligoclonally proliferated cells.

Fig. 6.

Comparison of TCR Vβ CDR3 complexity scores among CD8α+ αβ T cells with different CD8β expression.

Complexity scores were generated for each TCR BV from the CDR3 spectratype analysis. The individual complexity scores were plotted along CD8β expression, and the dots for the same BVs were connected with lines. Representative data of 2 donors are shown.

Fig. 6.

Comparison of TCR Vβ CDR3 complexity scores among CD8α+ αβ T cells with different CD8β expression.

Complexity scores were generated for each TCR BV from the CDR3 spectratype analysis. The individual complexity scores were plotted along CD8β expression, and the dots for the same BVs were connected with lines. Representative data of 2 donors are shown.

Close modal

Identical clones exist among CD8α+βhigh, CD8α+βlow, and CD8αα cells

It needs to be confirmed directly that CD8α+βlow and CD8αα αβ T cells are oligoclonally proliferated cells. Therefore, the PCR products were then cloned and the nucleotide sequence of CDR3 was determined (Table1). This analysis also provides the information if identical clones exist among the subpopulations of different CD8β expression. In this experiment, we used the cDNA samples from one donor so that the pruity of each sorted cell fraction was more than 98% and the number was identical for all BVs within a given cell fraction. In addition, we selected BV21, BV20, and BV14 because these BVs exhibited distinct patterns of spectratypes within CD8α+βhigh, CD8α+βlow, and CD8αα αβ T cells (BV21: polyclonal, polyclonal, and oligoclonal; BV20: polyclonal, oligoclonal, and oligoclonal; and BV14: oligoclonal, oligoclonal, and oligoclonal; Figure 7).

Table 1.

The amino acid composition of the CDR3 region within CD8α+βhigh, CD8α+βlow, and CD8αα αβ T cells

CD8+highCD8α+βlowCD8αα
BVN-D-NBJFrequencyBVN-D-NBJFrequencyBVN-D-NBJFrequency
BV21            
 CASS SGTAV SNQP (1S6) 2/19 CASS PVSGRL VNEQ (2S1) 3/16 CASS PVSGRL VNEQ (2S1) 9/16  
 CASS LDPSQGH TQVF (2S3) 2/19 CASS LDPSQGH TQVF (2S3) 3/16 CASS TTLAGT VNEQ (2S1) 1/16 
 CASS FVSGS STDT (2S3) 1/19 CASS FVSGS STDT (2S3) 3/16 CASS FVSGS STDT (2S3) 2/16 
 CASS TMG ETQV (2S5) 2/19 CASS PMR TDTQ (2S3) 1/16 CASS LEGRV QETQ (2S5) 1/16 
 CASS TPRTGS SGAN (2S6) 2/19 CASS PLSC EQFF (2S1) 1/16 CASS LVQGPP DTQV (2S3) 1/16 
 CASS PSL NTEA (1S1) 1/19 CASS SPGGA TQVF (2S3) 1/16 CASS LGGGSF VEQV (2S7) 1/16 
 CASS LGP NTEA (1S1) 1/19 CASS FVSVS STDT (2S3) 1/16 CAS RGLAA QETQ (2S5) 1/16 
 CASS SGTGALL EQFF (2S1) 1/19 CASS LGGGLSP KNIQ (2S4) 1/16 — — — — 
 CASS FPMGDRASGGD TGEL (2S2) 1/19 CASS SVCGRLS NEQF (2S1) 1/16 — — — — 
 CASS INPSQGH TQVF (2S3) 1/19 CASS FVSGRLSNA QVFG (2S3) 1/16 — — — — 
 CASS LAGGP TDTQ (2S3) 1/19 — — — — — — — — 
 CASS PSGTE ETQV (2S5) 1/19 — — — — — — — — 
 CASS LARD VEQV (2S7) 1/19 — — — — — — — — 
 CASS LGP SVEQ (2S7) 1/19 — — — — — — — — 
 CASS FGIGDS SVEQ (2S7) 1/19 — — — — — — — — 
BV20            
 CAW SPVSWA GNTI (1S3) 2/15 CAW SPVSWA GNTI (1S3) 9/11 CAW SPVSWA GNTI (1S3) 10/14 
 CA CRGCGR STDT (2S3) 1/15 CAWS GI VNEQ (2S1) 2/11 CAWS GI VNEQ (2S1) 1/14 
 CA QALI STDT (2S3) 1/15 — — — — CAW GLAD TDTQ (2S3) 1/14  
 CAW SAGTGG VEQV (2S7) 1/15 — — — — CAW SIGTSGM VEQV (2S1) 1/14  
 CAW SPSDGGRSLH NEQF (2S1) 1/15 — — — — CAWS FLSETGLI GELF (2S2) 1/14  
 CAW SDAGVH EQVF (2S7) 1/15 — — — — — — — — 
 CAW IRTSGAN NEQF (2S1) 1/15 — — — — — — — — 
 CAW MR DTQV (2S3) 1/15 — — — — — — — — 
 CAWS QGAGE EQFF (2S1) 1/15 — — — — — — — — 
 CAWS VLS TDTQ (2S3) 1/15 — — — — — — — — 
 CAWS VMAR VEQV (2S7) 1/15 — — — — — — — — 
 CAWS VEGV NEKL (1S4) 1/15 — — — — — — — — 
 CAWS VTGGQ DTQV (2S3) 1/15 — — — — — — — — 
 CAWS DPGD EQVF (2S7) 1/15 — — — — — — — — 
BV14    — — — — — — — — 
 ASSL GQSR ETQV (2S5) 7/11 ASSL GQSR ETQV (2S5) 10/11 ASSL GQSR ETQV (2S5) 6/13 
 ASS RGQG VEQV (2S7) 1/11 ASSL FPTGR EKLF (1S4) 1/11 ASSL FPTGR EKLF (1S4) 4/13 
 ASS SVGGRS EQFF (2S1) 1/11 — — — — ASSL EGQT SPLH (1S6) 2/13  
 ASS IAGIRTLD TDTQ (2S3) 1/11 — — — — ASS FELAGGA ETQV (2S5) 1/13  
 ASS SSGGS SVNE (2S1) 1/11 — — — — — — — — 
CD8+highCD8α+βlowCD8αα
BVN-D-NBJFrequencyBVN-D-NBJFrequencyBVN-D-NBJFrequency
BV21            
 CASS SGTAV SNQP (1S6) 2/19 CASS PVSGRL VNEQ (2S1) 3/16 CASS PVSGRL VNEQ (2S1) 9/16  
 CASS LDPSQGH TQVF (2S3) 2/19 CASS LDPSQGH TQVF (2S3) 3/16 CASS TTLAGT VNEQ (2S1) 1/16 
 CASS FVSGS STDT (2S3) 1/19 CASS FVSGS STDT (2S3) 3/16 CASS FVSGS STDT (2S3) 2/16 
 CASS TMG ETQV (2S5) 2/19 CASS PMR TDTQ (2S3) 1/16 CASS LEGRV QETQ (2S5) 1/16 
 CASS TPRTGS SGAN (2S6) 2/19 CASS PLSC EQFF (2S1) 1/16 CASS LVQGPP DTQV (2S3) 1/16 
 CASS PSL NTEA (1S1) 1/19 CASS SPGGA TQVF (2S3) 1/16 CASS LGGGSF VEQV (2S7) 1/16 
 CASS LGP NTEA (1S1) 1/19 CASS FVSVS STDT (2S3) 1/16 CAS RGLAA QETQ (2S5) 1/16 
 CASS SGTGALL EQFF (2S1) 1/19 CASS LGGGLSP KNIQ (2S4) 1/16 — — — — 
 CASS FPMGDRASGGD TGEL (2S2) 1/19 CASS SVCGRLS NEQF (2S1) 1/16 — — — — 
 CASS INPSQGH TQVF (2S3) 1/19 CASS FVSGRLSNA QVFG (2S3) 1/16 — — — — 
 CASS LAGGP TDTQ (2S3) 1/19 — — — — — — — — 
 CASS PSGTE ETQV (2S5) 1/19 — — — — — — — — 
 CASS LARD VEQV (2S7) 1/19 — — — — — — — — 
 CASS LGP SVEQ (2S7) 1/19 — — — — — — — — 
 CASS FGIGDS SVEQ (2S7) 1/19 — — — — — — — — 
BV20            
 CAW SPVSWA GNTI (1S3) 2/15 CAW SPVSWA GNTI (1S3) 9/11 CAW SPVSWA GNTI (1S3) 10/14 
 CA CRGCGR STDT (2S3) 1/15 CAWS GI VNEQ (2S1) 2/11 CAWS GI VNEQ (2S1) 1/14 
 CA QALI STDT (2S3) 1/15 — — — — CAW GLAD TDTQ (2S3) 1/14  
 CAW SAGTGG VEQV (2S7) 1/15 — — — — CAW SIGTSGM VEQV (2S1) 1/14  
 CAW SPSDGGRSLH NEQF (2S1) 1/15 — — — — CAWS FLSETGLI GELF (2S2) 1/14  
 CAW SDAGVH EQVF (2S7) 1/15 — — — — — — — — 
 CAW IRTSGAN NEQF (2S1) 1/15 — — — — — — — — 
 CAW MR DTQV (2S3) 1/15 — — — — — — — — 
 CAWS QGAGE EQFF (2S1) 1/15 — — — — — — — — 
 CAWS VLS TDTQ (2S3) 1/15 — — — — — — — — 
 CAWS VMAR VEQV (2S7) 1/15 — — — — — — — — 
 CAWS VEGV NEKL (1S4) 1/15 — — — — — — — — 
 CAWS VTGGQ DTQV (2S3) 1/15 — — — — — — — — 
 CAWS DPGD EQVF (2S7) 1/15 — — — — — — — — 
BV14    — — — — — — — — 
 ASSL GQSR ETQV (2S5) 7/11 ASSL GQSR ETQV (2S5) 10/11 ASSL GQSR ETQV (2S5) 6/13 
 ASS RGQG VEQV (2S7) 1/11 ASSL FPTGR EKLF (1S4) 1/11 ASSL FPTGR EKLF (1S4) 4/13 
 ASS SVGGRS EQFF (2S1) 1/11 — — — — ASSL EGQT SPLH (1S6) 2/13  
 ASS IAGIRTLD TDTQ (2S3) 1/11 — — — — ASS FELAGGA ETQV (2S5) 1/13  
 ASS SSGGS SVNE (2S1) 1/11 — — — — — — — — 

BV-Cβ amplification of sorted CD8α+βhigh, CD8α+βlow, and CD8αα αβ T cells in a healthy donor was pursued. BV21, BV20, and BV14 were selected for the sequence analysis using the same PCR products. Eleven to 19 randomly chosen clones were sequenced per each PCR product.

— indicates no data.

Fig. 7.

Spectratypes of TCR BV21, BV20, and BV14.

Spectratyping analysis of αβ T cells within CD8α+βhigh, CD8α+βlow, and CD8αα subpopulations was pursued in another healthy donor. Histograms of BV21, BV20, and BV14 are displayed as Figure 5.

Fig. 7.

Spectratypes of TCR BV21, BV20, and BV14.

Spectratyping analysis of αβ T cells within CD8α+βhigh, CD8α+βlow, and CD8αα subpopulations was pursued in another healthy donor. Histograms of BV21, BV20, and BV14 are displayed as Figure 5.

Close modal

As for BV21, 19 CDR3 cDNA clones of CD8α+βhigh cells were randomly selected and sequenced. Consistent with spectratyping, heterogeneous CDR3 clones were sequenced, which indicated that CD8α+βhigh cells possessing TCR Vβ21 were polyclonal. Conversely, a large number of cDNA clones were determined to be identical in CD8αα cells; in 9 of 16 clones the amino acid sequence of the N-D-N region was PVSGRL (designated as clone PVSGRL; single-letter amino acid codes). This clone was also identified in CD8α+βlow cells (3 of 16 clones). However, this clone PVSGRL was not found in the CD8α+βhigh subpopulation. Although an additional 46 cDNA clones within CD8α+βhighcells were analyzed, this clone was not detected (data not shown). In contrast, another clone, LDPSQGH, was detected within CD8α+βhigh cells and CD8α+βlow cells in the frequency of 2 of 19 and 3 of 16, respectively, but not within CD8αα cells. Notably, the third clone, FVSGS, was found within CD8α+βhigh, CD8α+βlow, and CD8αα cells, although the clone was not dominant within these subpopulations (1 of 19, 3 of 16, and 2 of 16, respectively).

In BV20, a major clone, SPVSWA, within CD8αα cells (10 of 14 clones) dominated within CD8α+βlow cells (9 of 11 clones). This clone was also detected within CD8α+βhigh cells (2 of 15 clones). In BV14, where spectratypes of CD8α+βhigh, CD8α+βlow, and CD8αα subpopulations were all oligoclonal, clone GQSR was identified predominantly within the cells of all the subpopulations. To ensure that sharing of the identical clones among CD8α+βhigh, CD8α+βlow, and CD8αα subpopulations holds true for other individuals, we determined CDR3 sequences of BV17 from a different healthy donor; a dominant clone, SATVSYEQY, (7 of 10 clones) and a clone KPAGTFVLF (2 of 10 clones) within CD8αα cells were also detected within CD8α+βhigh cells at a frequency of 3 of 18 and 1 of 18, respectively (data not shown). Taken together, it is proved that the cells with skewed BV spectratypes, frequently observed in CD8α+βlow and CD8αα subpopulations, comprise oligoclonally proliferated cells. More importantly, CD8α+βhigh, CD8α+βlow, and CD8αα αβ T cells can possess the same cell clones. Some of these clones also become dominant with the loss of CD8β chains. These results suggest that some cell clones proliferate while down-regulating CD8β chains.

Sj TREC concentrations decreased with the down-regulation of CD8β

If CD8αα αβ T cells descend from CD8α+βhigh αβ T cells, CD8αα cells have undergone cell division more than CD8α+βlow, and still more than CD8α+βhigh αβ T cells. To assess the relative proliferative history of CD8+ αβ T-cell populations defined by the intensity of CD8β expression, we measured Sj TREC concentrations in CD8α+βhigh, CD8α+βlow, and CD8αα αβ T-cell subsets. In all 3 donors examined, Sj TREC levels were higher in CD8α+βhigh αβ T cells, and the number of Sj TREC copies declined with the loss of CD8β expression (Table2). These results, supporting the findings of spectratyping analysis, indicate that CD8α+βhigh αβ T cells, at least at the population level, can differentiate to CD8αα αβ T cells but not the opposite way.

Table 2.

Quantification of Sj TREC within CD8α+βhigh, CD8α+βlow, and CD8αα αβ T cells

DonorSj TREC/5 × 104 cells
CD8α+βhighCD8α+βlowCD8αα
140 76 < 1 
87 32 < 1  
160 20 < 1 
DonorSj TREC/5 × 104 cells
CD8α+βhighCD8α+βlowCD8αα
140 76 < 1 
87 32 < 1  
160 20 < 1 

Sj TREC concentrations were analyzed in 3 independent healthy donors.

In the present paper, we tried to show that peripheral blood CD8αα or CD8α+βlow αβ T cells derive directly from CD8α+βhigh αβ T cells, presumably by an antigen-driven mechanism. Three different approaches were undertaken. At first, age-dependent changes in the proportions of CD8αα and CD8α+βlowαβ T cells were examined.

In cord blood, CD8αα αβ T cells were negligible and relatively small numbers of CD8α+βlow αβ T cells were detected. These populations increased with advancing age. They made up even about a half of the CD8α+ αβ T cells in some healthy adults. Neonatal T cells are exclusively composed of naive cells, and in repeated antigenic stimulations with age, infection and autoreactivity convert the naive cells to effector and memory cells.32 Therefore, CD8αα and CD8α+βlow αβ T cells may represent antigen-experienced cells. A wide range of the variation in the frequencies of these cells among adult individuals supports this notion.

Secondly, surface antigen expression, cytotoxic granule constituents, and cytokine production of CD8+ αβ T cells were assessed to characterize phenotype and functional features. The results of surface marker analysis were consistent with previous data that assessed surface expression of CD8αβ heterodimer on the positive and negative fractions of various antigens within CD8+ T cells.22 Several surface markers have been proposed for identification of naive, effector, and memory T cells. Although the CD45RA isoform can revert from CD45RO,33 double labeling with CD45RA and CD62L is frequently used for the identification of the cell phenotype.34,35 Alternatively, it is reported that simultaneous staining with CD45RA and CD27 mAbs can separate the majority of human CD8+ T cells into 3 functionally distinct subpopulations: CD45RA+ CD27+ cells, CD45RA+ CD27 cells, and CD45RACD27+ cells, which are naive, effector, and memory phenotype, respectively.36 Recently, naive T cells were identified as CD95 T lymphocytes.35 In addition, CCR7+ memory T cells were designated as central memory cells with the counterpart as effector memory cells.37 Considering data from these reports and that T cells reciprocally express CD45RA or CD45RO,38 the CD8α+βlow subpopulation contains effector memory cells. Although the CD8αα subpopulation exclusively comprised cells of the same surface phenotype as CD8α+βlow cells in more than half of the individuals analyzed, there were appreciable CD45ROCCR7 cells within the CD8αα subpopulation in others. Because of the skewed spectratypes and low Sj TREC concentration, these CD45ROCCR7 cells may represent terminally differentiated memory cells.39 

Perforin, granzyme B, and TIA-1 expression were compatible with the surface phenotype of CD8α+βlowsubpopulations because the cytotoxic granule constituents are effector cell properties.40 Perforin and TIA-1 were also produced in CD8αα cells. It remains to be elucidated that CD8αα cells did not express granzyme B. As for cytokine production, both CD8α+βlow and CD8αα cells produced IFN-γ but not IL-2. Naive cells solely produce IL-2, and effector cells generate IFN-γ but not IL-2. Memory cells, on the contrary, produce both of them.36,41 However, IL-2 production is prominent in central memory cells; that of effector memory cells is significantly reduced.37 Therefore, the results of the cytokine production also support that CD8α+βlow and CD8αα αβ T cells are effector memory cells.

Thirdly, analysis of CDR3 length diversity can be used to define the extent of clonal expansion within the TCR repertoire.42,43The TCR Vβ CDR3 complexity decreased in CD8+ T cells with diminished CD8β expression. The results suggest that CD8α+βlow and, moreover, CD8αα αβ T cells have proliferated extensively probably by antigenic stimulations. Also, the history of cell proliferation can be assessed by Sj TREC concentrations. Sj TRECs are the episomal DNA circles generated during excisional rearrangement of TCR genes.44 Not replicating during mitosis, they are diluted during cell division.45,46 Sj TREC value was significantly higher in CD8α+βhigh αβ T cells but reduced with the loss of CD8β chains, consistent with decrease in TCR Vβ CDR3 complexity. These data collectively indicated that CD8α+βhigh αβ T cells diminish the CD8β-chain expression as they are activated, proliferate, and acquire memory phenotypes and functions in vivo.

To prove directly that particular CD8+ T cells change the levels of β-chain expression in vivo, we tried to clarify that the same clones exist among CD8α+βhigh, CD8α+βlow, and CD8αα αβ T cells, and pursued TCR Vβ CDR3 sequence analysis. The CDR3 forms the contact site for binding to MHC expressed by antigen-presenting cells. The region bears the fine specificity of antigen recognition.47-50 Therefore, the CDR3 sequence defines a distinctive TCR clonotype that acts as a fingerprint for the T-cell lineage bearing it.51 On the basis of TCR Vβ CDR3 spectratyping and their sequences, recent studies proposed that identical T-cell clones are located within the mouse gut epithelium and lamina propria and circulate in the thoracic duct lymph,52or that CD8+CD28 T cells descend directly from CD8+CD28+ T cells in humans.53,54 In the experiment presented here, the same clones were identified among CD8α+βhigh, CD8α+βlow, and CD8αα αβ T cells in all of 3 BVs analyzed. Because the major clones sequenced within CD8αα cells were not identified within the CD8α+βhigh subset in BV21, it is unlikely that clones sequenced within the CD8α+βhighsubpopulation are derived from missorted CD8αα αβ T cells. This result may rather be explained by the fact that all cells of the clone have already down-regulated CD8β. Missorting can neither explain the result in BV14, which showed that the major clones sequenced among CD8α+βhigh, CD8α+βlow, and CD8αα αβ T cells were identical. Therefore, the sequence result suggests CD8α+βhigh, CD8α+βlow, and CD8αα αβ T cells can share the same clones. Considering the differentiation stage, spectratypes, Sj TREC concentration, and CDR3 sequences, CD8αα and CD8α+βlow αβ T cells do not belong to different ontogenic lineages, but directly descend from CD8α+βhigh αβ T cells.

In contrast to our data, numerous studies have suggested that CD8αα cells are extrathymically differentiated T cells.20-22,55CD8αα T cells reside in intestinal mucosa, and they are proved to be thymus independent.56-58 In bone marrow transplantation (BMT), about one half of the CD8+ T cells constituted from T cell–depleted marrow were CD8αα cells in thymectomized recipient mice.20 As for human children, older patients at BMT exhibited more CD8αα T cells than younger ones, suggesting the influence of thymic function on the regenerative pathways.21 However, these findings do not indicate that all of the CD8αα αβ T cells are necessarily of extrathymic origin. At least peripheral CD8αα αβ T cells and CD8αα αβ T intraepithelial lymphocytes (IELs) should be considered distinctively. It is reported that TCRαβ CD8αα IELs have not previously expressed CD8β using DNA methylation pattern analysis.59 Indeed, the results of IEL TCR Vβ CDR3 analysis turned out to be opposite to those presented in this study; TCRαβ IELs bearing CD8αβ or CD8αα coreceptors shared no TCR BV clone.52,60 

However, most recent studies by Leishman et al23 and Devine et al24 concerning the fate and function of these unique CD8+ T-cell subpopulations strongly indicate that they derive from the thymus and are positively selected. Moreover, CD8αα phenotype was shown to be acquired under the influence of the microenvironment of intraepithelial lymphoid tissue.25Furthermore, they suggest that CD8α+βlowand CD8αα αβ T cells exhibit unique functional roles in vivo distinct from CD8α+βhigh αβ T cells. Our findings are consistent with these data and confirm the notion that all 3 CD8+αβ T cells belong to the same cellular lineage and the level of CD8β chain expression is secondarily regulated in vivo by antigenic stimulation and subsequent cell activation.

In conclusion, the present study demonstrates that the CD8αα and CD8α+βlow αβ T cells are effector memory or terminally differentiated CD45ROCCR7 memory cells. Moreover, these cells share the same clone with the usual CD8α+βhigh αβ T cells. We propose that at least in healthy individuals, circulating CD8αα and CD8α+βlow αβ T cells do emerge as a consequence of MHC-TCR interaction. Therefore, the persistence of these cells reflects the presence of continuous antigenic stimulations and subsequent accumulation of the antigen-specific clonally expanded T cells, which may be influenced by thymic output. Analysis of these cells may help us to understand the pathogenesis of autoimmunity and immunodeficiency, and it will provide a useful biomarker to evaluate the disease activities.

We thank Ms Mika Kitakata, Ms Tamae Yonezawa, and Ms Harumi Matsukawa for excellent technical help.

Prepublished online as Blood First Edition Paper, July 25, 2002; DOI 10.1182/blood-2002-04-1136.

Supported by a Grant-in-Aid for Scientific Research from the Ministry of Education, Science, and Culture of Japan; and a grant from the Ministry of Health, Labour, and Welfare of Japan, Tokyo.

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.

1
Gao
GF
Tormo
J
Gerth
UC
et al
Crystal structure of the complex between human CD8alpha(alpha) and HLA-A2.
Nature.
387
1997
630
634
2
Zamoyska
R
CD4 and CD8: modulators of T-cell receptor recognition of antigen and of immune responses?
Curr Opin Immunol.
10
1998
82
87
3
Norment
AM
Salter
RD
Parham
P
Engelhard
VH
Littman
DR
Cell-cell adhesion mediated by CD8 and MHC class I molecules.
Nature.
336
1988
79
81
4
Salter
RD
Benjamin
RJ
Wesley
PK
et al
A binding site for the T-cell co-receptor CD8 on the alpha 3 domain of HLA-A2.
Nature.
345
1990
41
46
5
Veillette
A
Bookman
MA
Horak
EM
Bolen
JB
The CD4 and CD8 T cell surface antigens are associated with the internal membrane tyrosine-protein kinase p56lck.
Cell.
55
1988
301
308
6
Barber
EK
Dasgupta
JD
Schlossman
SF
Trevillyan
JM
Rudd
CE
The CD4 and CD8 antigens are coupled to a protein-tyrosine kinase (p56lck) that phosphorylates the CD3 complex.
Proc Natl Acad Sci U S A.
86
1989
3277
3281
7
Wange
RL
Samelson
LE
Complex complexes: signaling at the TCR.
Immunity.
5
1996
197
205
8
Baume
DM
Caligiuri
MA
Manley
TJ
Daley
JF
Ritz
J
Differential expression of CD8 alpha and CD8 beta associated with MHC-restricted and non-MHC-restricted cytolytic effector cells.
Cell Immunol.
131
1990
352
365
9
Moebius
U
Kober
G
Griscelli
AL
Hercend
T
Meuer
SC
Expression of different CD8 isoforms on distinct human lymphocyte subpopulations.
Eur J Immunol.
21
1991
1793
1800
10
Norment
AM
Littman
DR
A second subunit of CD8 is expressed in human T cells.
EMBO J.
7
1988
3433
3439
11
Terry
LA
DiSanto
JP
Small
TN
Flomenberg
N
Differential expression and regulation of the human CD8 alpha and CD8 beta chains.
Tissue Antigens.
35
1990
82
91
12
Nakayama
K
Shinkai
YI
Okumura
K
Nakauchi
H
Isolation and characterization of the mouse CD8 beta-chain (Ly-3) genes: absence of an intervening sequence between V- and J-like gene segments.
J Immunol.
142
1989
2540
2546
13
Parnes
JR
Molecular biology and function of CD4 and CD8.
Adv Immunol.
44
1989
265
311
14
Bosselut
R
Kubo
S
Guinter
T
et al
Role of CD8beta domains in CD8 coreceptor function: importance for MHC I binding, signaling, and positive selection of CD8+ T cells in the thymus.
Immunity.
12
2000
409
418
15
Renard
V
Romero
P
Vivier
E
Malissen
B
Luescher
IF
CD8 beta increases CD8 coreceptor function and participation in TCR-ligand binding.
J Exp Med.
184
1996
2439
2444
16
Wheeler
CJ
Chen
JY
Potter
TA
Parnes
JR
Mechanisms of CD8beta-mediated T cell response enhancement: interaction with MHC class I/beta2-microglobulin and functional coupling to TCR/CD3.
J Immunol.
160
1998
4199
4207
17
Irie
HY
Mong
MS
Itano
A
et al
The cytoplasmic domain of CD8 beta regulates Lck kinase activation and CD8 T cell development.
J Immunol.
161
1998
183
191
18
Irie
HY
Ravichandran
KS
Burakoff
SJ
CD8 beta chain influences CD8 alpha chain-associated Lck kinase activity.
J Exp Med.
181
1995
1267
1273
19
Kawabata
K
Nagasawa
M
Morio
T
Okawa
H
Yata
J
Decreased alpha/beta heterodimer among CD8 molecules of peripheral blood T cells in Wiskott-Aldrich syndrome.
Clin Immunol Immunopathol.
81
1996
129
135
20
Dulude
G
Brochu
S
Fontaine
P
et al
Thymic and extrathymic differentiation and expansion of T lymphocytes following bone marrow transplantation in irradiated recipients.
Exp Hematol.
25
1997
992
1004
21
Honda
K
Takada
H
Nagatoshi
Y
et al
Thymus-independent expansion of T lymphocytes in children after allogeneic bone marrow transplantation.
Bone Marrow Transplant.
25
2000
647
652
22
Schmitz
JE
Forman
MA
Lifton
MA
et al
Expression of the CD8alpha beta-heterodimer on CD8(+) T lymphocytes in peripheral blood lymphocytes of human immunodeficiency virus− and human immunodeficiency virus+ individuals.
Blood.
92
1998
198
206
23
Leishman
AJ
Gapin
L
Capone
M
et al
Precursors of functional MHC class I- or class II-restricted CD8aa T cells are positively selected in the thymus by agonist self-peptides.
Immunity.
16
2002
355
364
24
Devine
L
Rogozinski
L
Naidenko
OV
et al
The complementarity-determining region-like loops of CD8a interact differently with b2-microglobulin of the class I molecules H-2Kb and thymic leukemia antigen, while similarly with their a3 domains.
J Immunol.
168
2002
3881
3886
25
Imhof
BA
Dunon
D
Courtois
D
et al
Intestinal CD8aa and CD8ab intraepithelial lymphocytes are thymus derived and exhibit subtle differences in TCRb repertoires.
J Immunol.
165
2000
6716
6722
26
Hazenberg
MD
Otto
SA
Cohen Stuart
JW
et al
Increased cell division but not thymic dysfunction rapidly affects the T-cell receptor excision circle content of the naive T cell population in HIV-1 infection.
Nat Med.
6
2000
1036
1042
27
McFarland
RD
Douek
DC
Koup
RA
Picker
LJ
Identification of a human recent thymic emigrant phenotype.
Proc Natl Acad Sci U S A.
97
2000
4215
4220
28
Zeng
W
Nakao
S
Takamatsu
H
et al
Characterization of T-cell repertoire of the bone marrow in immune-mediated aplastic anemia: evidence for the involvement of antigen-driven T-cell response in cyclosporine-dependent aplastic anemia.
Blood.
93
1999
3008
3016
29
Choi
YW
Kotzin
B
Herron
L
Callahan
J
Marrack
P
Kappler
J
Interaction of Staphylococcus aureus toxin “superantigens” with human T cells.
Proc Natl Acad Sci U S A.
86
1989
8941
8945
30
Labrecque
N
McGrath
H
Subramanyam
M
Huber
BT
Sekaly
RP
Human T cells respond to mouse mammary tumor virus-encoded superantigen: V beta restriction and conserved evolutionary features.
J Exp Med.
177
1993
1735
1743
31
Bomberger
C
Singh-Jairam
M
Rodey
G
et al
Lymphoid reconstitution after autologous PBSC transplantation with FACS-sorted CD34+ hematopoietic progenitors.
Blood.
91
1998
2588
2600
32
Clement
LT
Isoforms of the CD45 common leukocyte antigen family: markers for human T-cell differentiation.
J Clin Immunol.
12
1992
1
10
33
Young
JL
Ramage
JM
Gaston
JS
Beverley
PC
In vitro responses of human CD45RObrightRA- and CD45RO-RAbright T cell subsets and their relationship to memory and naive T cells.
Eur J Immunol.
27
1997
2383
2390
34
Sempowski
G
Thomasch
J
Gooding
M
et al
Effect of thymectomy on human peripheral blood T cell pools in myasthenia gravis.
J Immunol.
166
2001
2808
2817
35
Fagnoni
FF
Vescovini
R
Passeri
G
et al
Shortage of circulating naive CD8(+) T cells provides new insights on immunodeficiency in aging.
Blood.
95
2000
2860
2868
36
Hamann
D
Baars
PA
Rep
MH
et al
Phenotypic and functional separation of memory and effector human CD8+ T cells.
J Exp Med.
186
1997
1407
1418
37
Sallusto
F
Lenig
D
Forster
R
Lipp
M
Lanzavecchia
A
Two subsets of memory T lymphocytes with distinct homing potentials and effector functions.
Nature.
401
1999
708
712
38
Richards
SJ
Jones
RA
Roberts
BE
Patel
D
Scott
CS
Relationships between 2H4 (CD45RA) and UCHL1 (CD45RO) expression by normal blood CD4+CD8−, CD4−CD8+, CD4−CD8dim+, CD3+CD4−CD8− and CD3−CD4−CD8− lymphocytes.
Clin Exp Immunol.
81
1990
149
155
39
Champagne
P
Ogg
GS
King
AS
et al
Skewed maturation of memory HIV-specific CD8 T lymphocytes.
Nature.
410
2001
106
111
40
Speiser
DE
Colonna
M
Ayyoub
M
et al
The activatory receptor 2B4 is expressed in vivo by human CD8(+) effector alphabeta T cells.
J Immunol.
167
2001
6165
6170
41
de Jong
R
Brouwer
M
Miedema
F
van Lier
RA
Human CD8+ T lymphocytes can be divided into CD45RA+ and CD45RO+ cells with different requirements for activation and differentiation.
J Immunol.
146
1991
2088
2094
42
Pannetier
C
Cochet
M
Darche
S
Casrouge
A
Zoller
M
Kourilsky
P
The sizes of the CDR3 hypervariable regions of the murine T-cell receptor beta chains vary as a function of the recombined germ-line segments.
Proc Natl Acad Sci U S A.
90
1993
4319
4323
43
Kou
ZC
Puhr
JS
Rojas
M
McCormack
WT
Goodenow
MM
Sleasman
JW
T-Cell receptor Vbeta repertoire CDR3 length diversity differs within CD45RA and CD45RO T-cell subsets in healthy and human immunodeficiency virus-infected children.
Clin Diagn Lab Immunol.
7
2000
953
959
44
Kong
F
Chen
CH
Cooper
MD
Thymic function can be accurately monitored by the level of recent T cell emigrants in the circulation.
Immunity.
8
1998
97
104
45
Livak
F
Schatz
DG
T-cell receptor alpha locus V(D)J recombination by-products are abundant in thymocytes and mature T cells.
Mol Cell Biol.
16
1996
609
618
46
Breit
TM
Verschuren
MC
Wolvers-Tettero
IL
Van Gastel-Mol
EJ
Hahlen
K
van Dongen
JJ
Human T cell leukemias with continuous V(D)J recombinase activity for TCR-delta gene deletion.
J Immunol.
159
1997
4341
4349
47
Wu
CJ
Chillemi
A
Alyea
EP
et al
Reconstitution of T-cell receptor repertoire diversity following T-cell depleted allogeneic bone marrow transplantation is related to hematopoietic chimerism.
Blood.
95
2000
352
359
48
Degano
M
Garcia
KC
Apostolopoulos
V
Rudolph
MG
Teyton
L
Wilson
IA
A functional hot spot for antigen recognition in a superagonist TCR/MHC complex.
Immunity.
12
2000
251
261
49
Jorgensen
JL
Esser
U
Fazekas de St Groth
B
Reay
PA
Davis
MM
Mapping T-cell receptor-peptide contacts by variant peptide immunization of single-chain transgenics.
Nature.
355
1992
224
230
50
Pannetier
C
Even
J
Kourilsky
P
T-cell repertoire diversity and clonal expansions in normal and clinical samples.
Immunol Today.
16
1995
176
181
51
Gorski
J
Yassai
M
Zhu
X
et al
Circulating T cell repertoire complexity in normal individuals and bone marrow recipients analyzed by CDR3 size spectratyping: correlation with immune status.
J Immunol.
152
1994
5109
5119
52
Arstila
T
Arstila
TP
Calbo
S
et al
Identical T cell clones are located within the mouse gut epithelium and lamina propria and circulate in the thoracic duct lymph.
J Exp Med.
191
2000
823
834
53
Mugnaini
EN
Egeland
T
Spurkland
A
Brinchmann
JE
The T cell receptor repertoire of CD8+CD28− T lymphocytes is dominated by expanded clones that persist over time.
Clin Exp Immunol.
117
1999
298
303
54
Horiuchi
T
Hirokawa
M
Kawabata
Y
et al
Identification of the T cell clones expanding within both CD8(+)CD28(+) and CD8(+)CD28(−) T cell subsets in recipients of allogeneic hematopoietic cell grafts and its implication in post-transplant skewing of T cell receptor repertoire.
Bone Marrow Transplant.
27
2001
731
739
55
Leclercq
G
Plum
J
Thymic and extrathymic T cell development.
Leukemia.
10
1996
1853
1859
56
Fung-Leung
WP
Kundig
TM
Ngo
K
et al
Reduced thymic maturation but normal effector function of CD8+ T cells in CD8 beta gene-targeted mice.
J Exp Med.
180
1994
959
967
57
Nakayama
K
Negishi
I
Kuida
K
et al
Requirement for CD8 beta chain in positive selection of CD8-lineage T cells.
Science.
263
1994
1131
1133
58
Rocha
B
Vassalli
P
Guy-Grand
D
Thymic and extrathymic origins of gut intraepithelial lymphocyte populations in mice.
J Exp Med.
180
1994
681
686
59
Hamerman
JA
Page
ST
Pullen
AM
Distinct methylation states of the CD8 beta gene in peripheral T cells and intraepithelial lymphocytes.
J Immunol.
159
1997
1240
1246
60
Regnault
A
Cumano
A
Vassalli
P
Guy-Grand
D
Kourilsky
P
Oligoclonal repertoire of the CD8 alpha alpha and the CD8 alpha beta TCR-alpha/beta murine intestinal intraepithelial T lymphocytes: evidence for the random emergence of T cells.
J Exp Med.
180
1994
1345
1358

Author notes

Akihiro Yachie, 13-1 Takara-machi, Kanazawa, 920-8641 Japan; e-mail:yachie@med.kanazawa-u.ac.jp.

Sign in via your Institution