Memory stem T cells (TSCMs) constitute a long-lived, self-renewing lymphocyte population essential for the maintenance of functional immunity. Hallmarks of autoimmune disease pathogenesis are abnormal CD4+ and CD8+ T cell activation. We investigated the TSCM subset in 55, 34, 43, and 5 patients with acquired aplastic anemia (AA), autoimmune uveitis, systemic lupus erythematosus, and sickle cell disease, respectively, as well as in 41 age-matched healthy controls. CD8+ TSCM frequency was significantly increased in AA compared with healthy controls. An increased CD8+ TSCM frequency at diagnosis was associated with responsiveness to immunosuppressive therapy, and an elevated CD8+ TSCM population after immunosuppressive therapy correlated with treatment failure or relapse in AA patients. IFN-γ and IL-2 production was significantly increased in various CD8+ and CD4+ T cell subsets in AA patients, including CD8+ and CD4+ TSCMs. CD8+ TSCM frequency was also increased in patients with autoimmune uveitis or sickle cell disease. A positive correlation between CD4+ and CD8+ TSCM frequencies was found in AA, autoimmune uveitis, and systemic lupus erythematosus. Evaluation of PD-1, CD160, and CD244 expression revealed that TSCMs were less exhausted compared with other types of memory T cells. Our results suggest that the CD8+ TSCM subset is a novel biomarker and a potential therapeutic target for AA.

Aplastic anemia (AA), the prototypical bone marrow (BM) failure syndrome, is caused by immune-mediated destruction of hematopoietic stem/progenitor cells (HSPCs) (1). BM transplantation and immunosuppressive therapy (IST) are effective for the treatment of AA (2). Although no Ag has been convincingly demonstrated in AA, autoimmunity to HSPCs has been considered etiologic, mediated by activated CTLs recognizing an HSPC-restricted Ag through their class I or II HLA molecules (3, 4). CD8+ CTLs with restricted TCR diversity (oligoclonal T cells) are expanded in AA, leading to the production of proinflammatory cytokines (e.g., IFN-γ) that induces apoptosis of CD34+ cells (5). Detection of similar clonotypes, as shown by significantly skewed CDR3 size distribution, suggests the presence of common Ag-driven T cell expansion (6). In AA, there is overrepresentation of HLA-DR2 and class I HLA-A*02:01, -A*02:06, -A*31:01, and -B*40:02 (7, 8). Hematologic recovery after IST with anti-thymocyte globulin (ATG) and cyclosporine (CsA) occurs in 60–75% of AA patients (2, 9), which correlates with diminution of expanded T cell clones. Relapse appears to be associated with re-emergence of the original oligoclonal T cells and, sometimes, new clones (6).

Conventionally, memory T cells are divided into central memory T cell (TCM) and effector memory T cell (TEM) subsets that home to secondary lymphoid and peripheral tissues, respectively (10). The percentages of CD4+ and CD8+ TEM subsets are increased in peripheral blood (PB) and BM of AA patients (11). Elevated TEM cells with potent effector capacity may relate to abnormal immunity in AA. Recent studies identified a new subset of memory stem T cells (TSCMs) (12, 13), which are the least differentiated cells of all distinct memory populations. These TSCMs express multiple naive markers, as well as the memory Ag CD95. Functionally, they possess an enhanced capacity for self-renewal, can generate multiple memory T cell populations, and likely play important roles in controlling immunity (12). In addition, these TSCMs are endowed with superior immune reconstitution potential in immunodeficient hosts and can mediate antitumor immunity in a humanized mouse model (12).

In autoimmune diseases, there is abnormal CD4+ and CD8+ T cell activation. We hypothesized dysregulation of the TSCM compartment in autoimmunity. In this study, we evaluated TSCM frequency in AA and its association with severity, treatment response, relapse, and changes after IST. Further, to evaluate TSCMs in other autoimmune diseases, we examined CD4+ and CD8+ TSCM frequencies in uveitis, systemic lupus erythematosus (SLE), and sickle cell disease (SCD) compared with healthy controls. Our results suggest that TSCMs may contribute to the pathophysiology in cancer immunology, as well as in autoimmune diseases. Specifically, CD8+ TSCMs may be a biomarker and a therapeutic target in AA.

PB specimens were collected from 55 AA samples and 41 age-matched healthy donor samples after informed consent was obtained in accordance with the Declaration of Helsinki. Of the 55 AA samples, 21 were analyzed at diagnosis, and 34 were analyzed after IST. Standard criteria were used for diagnosis and disease severity of AA (14, 15). Serial samples were collected before and after IST from 13 patients. The median age of the AA patients was 29 y (range: 13–69 y). All AA patients received horse ATG + CsA + eltrombopag in a clinical research protocol (clinicaltrials.gov, #NCT01623167). For comparison, blood samples were obtained from 34 uveitis patients (27 inactive and 7 active cases), 43 SLE patients who met the American College of Rheumatology criteria for the disease (16, 17) (19 with inactive SLE [SLE disease activity index-2K (SLEDAI-2K) score ≤ 3] and 24 with active SLE [SLEDAI-2K score > 3]), and 5 SCD patients who were receiving frequent transfusions. Demographic and clinical characteristics of patients and healthy controls are summarized in Table I. There were no significant differences in the median age between each patient group and healthy controls (p > 0.05, Supplemental Fig. 1A). All human subjects were enrolled in clinical protocols approved by the National Heart, Lung, and Blood Institute, National Eye Institute, and National Institute of Arthritis and Musculoskeletal and Skin Diseases Institutional Review Boards.

Table I.
Characteristics of patients and healthy controls
CharacteristicAA (n = 55)aUveitis (n = 34)SLE (n = 43)SCD (n = 5)HC (n = 41)
Median age (y) 29 55 48 50 33 
Sex (n; male/female) 33/22 11/23 5/38 2/3 21/20 
Diagnosis (nIdiopathic (55) Sarcoidosis (8) Idiopathic (43) Sickle cell anemia (5) NA 
  VKH (9)    
  Birdshot (15)    
  Idiopathic (2)    
Disease activity or severity (nSevere (55) Active (7) SLEDAI-2K > 3 (24) NA NA 
 Nonsevere (0) Quiet (27) SLEDAI-2K ≤ 3 (19)   
Time of sampling (n     
 Before therapy 21 NA 
 After therapy 34 25 42  
Therapy (nATG+CsA+TPO-RA (55) PSL (11) PSL (33) Transfusion program (4) NA 
  MMF (14) HCQ (33) Transfusion + HU (1)  
  CsA (4) MMF (6)   
  MTX (6) AZA (9)   
  Anti-TNF (6) MTX (10)   
CharacteristicAA (n = 55)aUveitis (n = 34)SLE (n = 43)SCD (n = 5)HC (n = 41)
Median age (y) 29 55 48 50 33 
Sex (n; male/female) 33/22 11/23 5/38 2/3 21/20 
Diagnosis (nIdiopathic (55) Sarcoidosis (8) Idiopathic (43) Sickle cell anemia (5) NA 
  VKH (9)    
  Birdshot (15)    
  Idiopathic (2)    
Disease activity or severity (nSevere (55) Active (7) SLEDAI-2K > 3 (24) NA NA 
 Nonsevere (0) Quiet (27) SLEDAI-2K ≤ 3 (19)   
Time of sampling (n     
 Before therapy 21 NA 
 After therapy 34 25 42  
Therapy (nATG+CsA+TPO-RA (55) PSL (11) PSL (33) Transfusion program (4) NA 
  MMF (14) HCQ (33) Transfusion + HU (1)  
  CsA (4) MMF (6)   
  MTX (6) AZA (9)   
  Anti-TNF (6) MTX (10)   
a

Because serial samples were collected from 13 patients before and after IST (including 3 patients at three time points), the actual patient number is 39.

AZA, azathioprine; HC, healthy controls; HU, hydroxyurea; NA, not applicable; TPO-RA, thrombopoietin receptor agonist; VKH, Vogt–Koyanagi–Harada disease.

PBMCs were separated from PB samples using Lymphocyte Separation Medium (MP Biomedicals, Solon, OH) and cryopreserved in RPMI 1640 (Life Technologies, Gaithersburg, MD) supplemented with 20% heat-inactivated FBS (Sigma-Aldrich, St Louis, MO) and 10% DMSO, according to the standard protocol, until use.

The following fluorochrome-conjugated mAbs were purchased from commercial vendors and used for surface staining: anti-CD4–V500, anti-CD8–allophycocyanin–H7, anti-CD45RA–PE–Cy7, anti-CD45RO–allophycocyanin, anti-CCR7–Alexa Fluor 700, anti-CD95–PE, anti-CD160–FITC, anti-CD244–FITC, and anti-PD-1–FITC (all from BD Biosciences, San Jose, CA); anti-CD3–BV605 (BioLegend, San Diego, CA); anti-CD14–Pacific Blue and anti-CD19–Pacific Blue (Life Technologies, Carlsbad, CA); and anti-CD27–PC5 (Beckman Coulter, Indianapolis, IN). The fixable violet amine reactive dye (ViViD; Invitrogen/Molecular Probes, Eugene, OR) was used to eliminate dead cells by flow cytometry. For intracellular cytokine staining, the following mAbs were used: anti-granzyme B (GZMB)-FITC, anti-IL-2–FITC, and anti-IFN-γ–FITC (BD Biosciences).

The gating strategy for TSCMs and experimental protocols were adapted from a previous article (18). Briefly, cryopreserved PBMCs were thawed and subjected to surface staining as follows. PBMCs were incubated with a viability marker at room temperature for 20 min, washed, and incubated with anti-human CCR7 at 37°C for 20 min. After washing, cells were stained with a mixture of Abs against CD3, CD4, CD8, CD45RA, CD45RO, CD27, CD95, CD14, and CD19, with or without exhaustion markers (anti–PD-1 Ab, anti-CD160 Ab, and anti-CD244 Ab, respectively), for 30 min on ice. Subsequently, cells were washed, and ≥150,000 events gated on CD3+ T cells were acquired with a Fortessa flow cytometer (BD Biosciences) to analyze the frequency of each T cell subset. The gating strategy for T cell subsets is summarized in Fig. 1A. Lymphocytes were gated based on their scatter characteristics, and a forward scatter-area versus forward scatter-height profile was used to exclude cell aggregates and to obtain single lymphocytes. Live T cells were separated from dead cells, monocytes, and B cells in a CD3 versus ViViD/CD14/CD19 bivariate plot. CD4+ and CD8+ T cells were then gated based on characteristic expression patterns of CCR7 and CD45RO, followed by gating based on CD27/CD45RA and CCR7/CD95 expression. Each T cell subset was defined as follows: TCMs: ViViD CD3+ CD4 (CD8)+ CD45RO+ CCR7+; TEMs: ViViD CD3+ CD4 (CD8)+ CD45RO+ CCR7; terminally differentiated TEMs (TEs): ViViD CD3+ CD4 (CD8)+ CD45RO CD45RA+ CCR7 CD27; naive T cells (TNs): CD3+ CD4 (CD8)+ CD45RO CD45RA+ CCR7+ CD27+ CD95; and TSCMs, CD3+ CD4 (CD8)+ CD45RO CD45RA+ CCR7+ CD27+ CD95+. Data were analyzed using FlowJo software version 9.6 (TreeStar, Ashland, OR). The quantification of inhibitory receptor expression in T cell subsets was described in detail (19).

FIGURE 1.

The increased CD8+ TSCM population in AA patients. (A) Gating strategy for T cell subsets. PBMCs were stained with ViViD, anti-CD14–Pacific Blue, anti-CD19–Pacific Blue, anti-CD3–BV605, anti-CD4–V500, anti-CD8–allophycocyanin–H7, anti-CD45RA–PE–Cy7, anti-CD45RO–allophycocyanin, anti-CCR7–Alexa Fluor 700, anti-CD27–PC5, and anti-CD95–PE. Lymphocytes or single lymphocytes were gated based on their scatter characteristics or forward scatter height versus forward scatter area, respectively. Live T cells were gated based on positivity for CD3 and negativity for ViViD, CD14, and CD19 to remove dead cells, monocytes, and B cells. CD4+ and CD8+ T cells were then gated based on the characteristic expression patterns of CCR7 and CD45RO. ViViD CD3+ CD4 (CD8)+ CD45RO CD45RA+ CCR7+ CD27+ CD95 TNs, ViViD CD3+ CD4 (CD8)+ CD45RO CD45RA+ CCR7+ CD27+ CD95+ TSCMs, ViViD CD3+ CD4 (CD8)+ CD45RO+ CCR7+ TCMs, ViViD CD3+ CD4 (CD8)+ CD45RO+ CCR7 TEMs, and ViViD CD3+ CD4 (CD8)+ CD45RO CD45RA+ CCR7 CD27 TEs were identified. (B) Frequency of each CD4+ or CD8+ T cell subset (TN, TSCM, TCM, TEM, or TE) was compared between AA (n = 55) and healthy control (n = 41) groups. (C) Frequencies of CD4+ and CD8+ TSCM populations were compared within the same group (AA [n = 55] or healthy control group [n = 41]) or between the two groups. (D) Representative flow cytometry dot plots illustrate the increased CD8+ TSCM population in an AA patient (left panel) relative to a healthy individual (right panel). *p < 0.05, Student t test.

FIGURE 1.

The increased CD8+ TSCM population in AA patients. (A) Gating strategy for T cell subsets. PBMCs were stained with ViViD, anti-CD14–Pacific Blue, anti-CD19–Pacific Blue, anti-CD3–BV605, anti-CD4–V500, anti-CD8–allophycocyanin–H7, anti-CD45RA–PE–Cy7, anti-CD45RO–allophycocyanin, anti-CCR7–Alexa Fluor 700, anti-CD27–PC5, and anti-CD95–PE. Lymphocytes or single lymphocytes were gated based on their scatter characteristics or forward scatter height versus forward scatter area, respectively. Live T cells were gated based on positivity for CD3 and negativity for ViViD, CD14, and CD19 to remove dead cells, monocytes, and B cells. CD4+ and CD8+ T cells were then gated based on the characteristic expression patterns of CCR7 and CD45RO. ViViD CD3+ CD4 (CD8)+ CD45RO CD45RA+ CCR7+ CD27+ CD95 TNs, ViViD CD3+ CD4 (CD8)+ CD45RO CD45RA+ CCR7+ CD27+ CD95+ TSCMs, ViViD CD3+ CD4 (CD8)+ CD45RO+ CCR7+ TCMs, ViViD CD3+ CD4 (CD8)+ CD45RO+ CCR7 TEMs, and ViViD CD3+ CD4 (CD8)+ CD45RO CD45RA+ CCR7 CD27 TEs were identified. (B) Frequency of each CD4+ or CD8+ T cell subset (TN, TSCM, TCM, TEM, or TE) was compared between AA (n = 55) and healthy control (n = 41) groups. (C) Frequencies of CD4+ and CD8+ TSCM populations were compared within the same group (AA [n = 55] or healthy control group [n = 41]) or between the two groups. (D) Representative flow cytometry dot plots illustrate the increased CD8+ TSCM population in an AA patient (left panel) relative to a healthy individual (right panel). *p < 0.05, Student t test.

Close modal

Expression levels of GZMB, IL-2, and IFN-γ in CD4+ and CD8+ T cell subsets were analyzed by intracellular cytokine staining 6 h poststimulation. Briefly, cells were stimulated by addition of Dynabeads Human T-Activator CD3/CD28 and then 2 h later by the addition of Golgi transport inhibitor (GolgiPlug; BD Biosciences). After another 4 h of culture, cells were incubated with the cell surface–staining Ab mixture, as described in Immunostaining for surface Ags, and were fixed/permeabilized using Cytofix/Cytoperm Fixation and Permeabilization Solution (BD Biosciences), according to the manufacturer’s protocol. Subsequently, intracellular cytokine staining was performed using anti-GZMB–FITC, anti-IL-2–FITC, and anti-IFN-γ–FITC at 4°C for 30 min.

All statistical analyses were performed using GraphPad PRISM version 6.0 (GraphPad, La Jolla, CA). Data are shown as mean ± SEM. A Student t test was used to calculate statistical significance between two groups. A statistical analysis was performed using one-way or two-way ANOVA with the post hoc Tukey or Dunnett test for multiple comparisons, when appropriate. The Spearman rank test with linear regression was used for correlation analysis. A two-tailed p value < 0.05 was considered statistically significant.

First, we measured five T cell subsets (TN, TSCM, TCM, TEM, and TE) in AA and healthy controls. Within the CD4+ or CD8+ T cell compartments, AA patients showed decreased CD4+ or CD8+ TN frequency (p < 0.05, Fig. 1B), compared with controls, consistent with previous reports (11). CD4+ TE frequency was very low in the CD4+ T cell compartment in both AA and controls, but CD8+ TE frequency was higher among CD8+ T cells in both. In healthy controls, TSCMs represented a relatively small percentage of circulating CD4+ or CD8+ T cells (median 2.4% CD4+ TSCMs and 2.1% CD8+ TSCMs), confirming the findings of Gattinoni et al. (12). Samples collected from the same healthy donors but on different dates showed similar results, confirming technical and biological reproducibility (Supplemental Fig. 2). A significantly higher CD8+ TSCM frequency was detected in AA patients (4.2% versus 2.1%, p < 0.05), whereas there was no difference in CD4+ TSCM frequency (p > 0.05) compared with controls (Fig. 1C, 1D). Within the AA group, CD8+ TSCMs (4.2%) were more frequent than CD4+ TSCMs (2.1%) (p < 0.05, Fig. 1C), whereas the frequencies of CD4+ and CD8+ TSCMs within the control group showed no differences.

We assessed TSCM subset correlations with clinical manifestations and treatment responses in the AA cohort. CD4+ and CD8+ TSCM populations were evaluated in AA patients by clinical parameters, including age, sex, severity, and responses to IST. Responses to IST were defined according to established criteria (20). In AA (n = 21), CD8+ TSCM frequency was measured at diagnosis, and response was assessed at 3 mo post-IST (Fig. 2A). In AA, a high CD8+ TSCM frequency at diagnosis correlated with a complete response (CR) or partial response (PR) to IST (5.0% in CR and PR versus 2.8% in nonresponders [NRs], p < 0.05) (Fig. 2A). In AA patients prior to IST (n = 21), CD8+ TSCM frequency did not correlate with age, sex, absolute neutrophil count, platelet count, time from diagnosis to therapy, or serum ferritin levels (p > 0.05, Supplemental Fig. 3A). To elucidate the effects of IST on the TSCM populations in AA patients, we next compared CD4+ or CD8+ TSCM frequency among three groups: 21 AA subjects without IST, 34 AA subjects with IST (3 mo to 2 y post-IST), and 41 healthy controls. CD8+ TSCMs were significantly increased in the two AA cohorts (with or without IST) relative to controls (p < 0.05), but CD4+ TSCM frequency was not statistically different among the three groups (p > 0.05) (Fig. 2B). We further evaluated CD4+ or CD8+ TSCM frequency among three cohorts (27 responders [CR or PR], 7 NRs or relapsed cases after IST, and 41 healthy donors). CD4+ TSCM frequency was not significantly different, but a higher CD8+ TSCM frequency was observed in the 7 NRs or relapsed cases after IST compared with the 27 responders (CR or PR) and with the 41 controls. A higher CD8+ TSCM frequency after IST was associated with treatment failure (3.5% in responders versus 5.5% in NRs or relapse, p < 0.05) (Fig. 2C). In serially collected samples from 13 AA patients, CD8+ TSCM frequency tended to decrease or remain the same after IST, with the exception of two patients (relapse at 9 mo and NR at 3 mo) who showed marked expanded CD8+ TSCMs after IST (Fig. 2D).

FIGURE 2.

Clinical correlations with the TSCM populations in AA patients. (A) CD8+ TSCM frequency in AA patients was measured at diagnosis (y-axis) and then examined at 3 mo post-IST (x-axis): CR and PR patients were combined into one group, and NR patients were combined into the other group. (B) CD4+ or CD8+ TSCM frequency was compared among three groups (AA patients without IST [n = 21] and with IST [n = 34] and healthy controls [n = 41]). (C) Frequency comparison of CD4+ or CD8+ TSCMs was performed among three groups: CR or PR after IST (n = 27), NR or relapse after IST (n = 7), and healthy controls (n = 41). (D) CD8+ TSCM frequency was measured at diagnosis and then after IST at different time points in the same 13 AA patients (left panel). Representative flow cytometry dot plots depict CD8+ TSCM frequency in one AA patient at diagnosis and relapse at 9 mo after IST (right panel). *p < 0.05, Student t test.

FIGURE 2.

Clinical correlations with the TSCM populations in AA patients. (A) CD8+ TSCM frequency in AA patients was measured at diagnosis (y-axis) and then examined at 3 mo post-IST (x-axis): CR and PR patients were combined into one group, and NR patients were combined into the other group. (B) CD4+ or CD8+ TSCM frequency was compared among three groups (AA patients without IST [n = 21] and with IST [n = 34] and healthy controls [n = 41]). (C) Frequency comparison of CD4+ or CD8+ TSCMs was performed among three groups: CR or PR after IST (n = 27), NR or relapse after IST (n = 7), and healthy controls (n = 41). (D) CD8+ TSCM frequency was measured at diagnosis and then after IST at different time points in the same 13 AA patients (left panel). Representative flow cytometry dot plots depict CD8+ TSCM frequency in one AA patient at diagnosis and relapse at 9 mo after IST (right panel). *p < 0.05, Student t test.

Close modal

In AA patients, a high CD8+ TSCM frequency at diagnosis correlated with better response to IST, whereas an increased CD8+ TSCM frequency was observed in NRs or relapsed cases after IST, suggesting TSCM as a potential biomarker. Memory T cell subsets often are described by their effector functions: TCMs are distinguished by greater proliferative and IL-2–producing capacities, and TEMs are characterized by increased secretion of effector cytokines, such as IFN-γ and GZMB. Stimulation with anti-CD3/CD28 beads successfully induced cytokine production by CD4+ and CD8+ T cells from AA and healthy controls (Fig. 3A). Elevated IFN-γ and IL-2 levels were observed in both total CD4+ and CD8+ T cell populations in AA patients compared with healthy controls (Fig. 3B, 3C). We next investigated which CD4+ or CD8+ T cell subsets (TNs, TSCMs, TCMs, TEMs, and TEs) were responsible for cytokine (GZMB, IFN-γ, and IL-2) production (Fig. 3B, 3C). Consistent with observations in total CD4+ and CD8+ T cell populations, no significant differences were detected in the frequencies of GZMB-producing CD4+ and CD8+ T cell subsets in AA patients compared with normal controls (Fig. 3B, 3C). In the CD4+ T cell compartment, IFN-γ and IL-2 levels were significantly elevated in the TSCM, TCM, and TEM subsets in AA. T cell subsets in the CD8+ T cell compartment showed greatly increased IFN-γ levels in CD8+ TSCM, TCM, TEM, and TE subsets and moderately elevated IL-2 levels in the CD8+ TSCM and TCM subsets. Taken together, elevated IFN-γ and IL-2 levels were seen in CD4+ and CD8+ TSCMs in AA compared with healthy controls (Fig. 3B, 3C).

FIGURE 3.

Cytokine production in CD4+ and CD8+ TSCM populations in AA patients. Cytokine production (GZMB, IFN-γ, and IL-2) was induced by in vitro stimulation with anti-CD3/CD28 beads, followed by immunostaining for intracellular cytokines. (A) Representative graphs showing cytokine production (IFN-γ and IL-2) of CD4+ and CD8+ T cell subsets in AA patients and healthy controls. A percentage of cytokine-producing (GZMB, IFN-γ, or IL-2) CD4+ (B) or CD8+ (C) T cell subsets was compared between AA patients at diagnosis (n = 4) and healthy controls (n = 5). *p < 0.05.

FIGURE 3.

Cytokine production in CD4+ and CD8+ TSCM populations in AA patients. Cytokine production (GZMB, IFN-γ, and IL-2) was induced by in vitro stimulation with anti-CD3/CD28 beads, followed by immunostaining for intracellular cytokines. (A) Representative graphs showing cytokine production (IFN-γ and IL-2) of CD4+ and CD8+ T cell subsets in AA patients and healthy controls. A percentage of cytokine-producing (GZMB, IFN-γ, or IL-2) CD4+ (B) or CD8+ (C) T cell subsets was compared between AA patients at diagnosis (n = 4) and healthy controls (n = 5). *p < 0.05.

Close modal

We next compared CD4+ or CD8+ TSCM frequency between each patient group (AA, uveitis, SLE, or SCD) and a healthy control group (Table I). We chose uveitis as an organ-specific immune-mediated disorder characterized by inflammatory ocular lesions, SLE as a systemic autoimmune disease, and SCD as controls for transfusions. Among the four patient groups, the uveitis group displayed a reduction in CD4+ TSCM frequency (1.8%) relative to the healthy controls (2.4%, p < 0.05). An elevated CD8+ TSCM frequency was observed in AA (4.2%), uveitis (3.6%), and SCD (4.3%), but not in SLE, compared with controls (2.1%, p < 0.05) (Fig. 4A). CD4+ and CD8+ TSCM frequencies were positively correlated in patients with AA (r = +0.45, p = 0.0010), uveitis (r = +0.67, p < 0.0001), or SLE (r = +0.47, p = 0.0024) but not in healthy controls (r = +0.14, p = 0.42) (Fig. 4B). CD4+ TSCM frequency was inversely correlated with TEM frequency in AA (r = −0.38, p = 0.0045), uveitis (r = −0.58, p = 0.0003), and SLE (r = −0.49, p = 0.0009) (Supplemental Fig. 4A). For CD8+ TSCM and TE frequencies, the same inverse correlation was also observed in AA (r = −0.39, p = 0.0044) and uveitis (r = −0.55, p = 0.0010) (Supplemental Fig. 4B). There was no correlation between TSCM frequency and age (r = +0.03, p = 0.70 for CD4+ TSCMs; r = +0.06, p = 0.41 for CD8+ TSCMs, Supplemental Fig. 1B, 1C).

FIGURE 4.

Comparison of CD4+ or CD8+ TSCM populations within the AA, uveitis, SLE, or SCD cohorts. (A) CD4+ or CD8+ TSCM frequency was compared among AA (n = 55), uveitis (n = 34), SLE (n = 43), SCD (n = 5), and healthy control (n = 41) cohorts. (B) Results of correlation Spearman rank tests in which CD4+ TSCM frequency was compared with CD8+ TSCM frequency within the same group (AA, uveitis, SLE, or healthy control). *p < 0.05.

FIGURE 4.

Comparison of CD4+ or CD8+ TSCM populations within the AA, uveitis, SLE, or SCD cohorts. (A) CD4+ or CD8+ TSCM frequency was compared among AA (n = 55), uveitis (n = 34), SLE (n = 43), SCD (n = 5), and healthy control (n = 41) cohorts. (B) Results of correlation Spearman rank tests in which CD4+ TSCM frequency was compared with CD8+ TSCM frequency within the same group (AA, uveitis, SLE, or healthy control). *p < 0.05.

Close modal

Aberrant overexpression of PD-1 (a cell surface inhibitory receptor, also known as an exhaustion marker of T cells) is associated with persistent activation of self-reactive T cells in autoimmune diseases (2123). We measured surface PD-1 expression on individual CD4+ or CD8+ T subsets (focusing on TSCMs) in four groups (AA, uveitis, SLE, and healthy control) (Fig. 5A). In CD4+ T cell subsets, PD-1 expression was lower on TSCMs than on TCMs, TEMs, and TEs within clinical groups (AA, uveitis, SLE, and healthy controls, p < 0.05), identifying TSCMs as the least exhausted population among CD4+ T cells. When PD-1 expression levels on individual CD4+ T cell subsets were compared between each patient group and controls, significantly higher PD-1 expression levels were seen on TEs in uveitis and on TCMs, TEMs, and TEs in SLE (p < 0.05, Fig. 5B). In CD8+ T cell subsets, PD-1 expression also was significantly lower on TSCMs than on TCMs and TEMs in all four groups (p < 0.05) (Fig. 5B). Significantly higher PD-1 expression levels were seen on CD8+ TEMs in AA. These results were consistent with previous reports demonstrating that PD-1 expression correlated with the differentiation of CD4+ and CD8+ T cells (24, 25). When PD-1 expression on CD4+ or CD8+ TSCMs was compared among AA, uveitis, SLE, and controls, it was elevated on CD4+ TSCMs in SLE and on CD8+ TSCMs in AA and uveitis (p < 0.05, Fig. 5C).

FIGURE 5.

Inhibitory receptor expression on T cell subsets. (A) Representative graphs of inhibitory receptor (PD-1, CD160, and CD244) expression of CD4+ and CD8+ T cell subsets in AA, uveitis, SLE, and healthy controls. (B) Surface inhibitory receptor (PD-1, CD160, and CD244) expression levels on CD4+ and CD8+ T cell subsets were analyzed in AA (n = 16), uveitis (n = 15), SLE (n = 22), and healthy controls (n = 9) and compared within the same groups or between each patient and control groups. Horizontal lines indicate the statistically significant changes (p < 0.05). (C) PD-1, CD160, and CD244 expression of CD4+ and CD8+ TSCMs plotted using a small-scale range to highlight differences in their values. They were compared between each patient and control groups. Boxes represent median and 25th and 75th percentiles; whiskers represent 10th and 90th percentiles. *p < 0.05.

FIGURE 5.

Inhibitory receptor expression on T cell subsets. (A) Representative graphs of inhibitory receptor (PD-1, CD160, and CD244) expression of CD4+ and CD8+ T cell subsets in AA, uveitis, SLE, and healthy controls. (B) Surface inhibitory receptor (PD-1, CD160, and CD244) expression levels on CD4+ and CD8+ T cell subsets were analyzed in AA (n = 16), uveitis (n = 15), SLE (n = 22), and healthy controls (n = 9) and compared within the same groups or between each patient and control groups. Horizontal lines indicate the statistically significant changes (p < 0.05). (C) PD-1, CD160, and CD244 expression of CD4+ and CD8+ TSCMs plotted using a small-scale range to highlight differences in their values. They were compared between each patient and control groups. Boxes represent median and 25th and 75th percentiles; whiskers represent 10th and 90th percentiles. *p < 0.05.

Close modal

To verify the results for PD-1 expression in T cell subsets, the expression of other inhibitory receptors (CD160 and CD244) was examined in T cell subsets from various autoimmune diseases (19, 26, 27). CD160 and CD244 were primarily expressed in CD8+ T cell subsets, among which the CD8+ TSCM population was less exhausted compared with the other T cell subsets (Fig. 5A, 5B). CD160, as well as PD-1, was highly expressed in CD8+ TSCMs of AA compared with healthy donors (Fig. 5C).

Finally, we assessed TSCM subset correlations with clinical manifestations and treatment responses in the uveitis and SLE cohorts. CD4+ and CD8+ TSCM populations were evaluated in patients with or without some clinical parameters, including age, sex, severity, and immune therapies specific for individual diseases. In uveitis, patients (n = 34) were classified into two groups, with or without current IST, and CD8+ TSCM frequency was compared between the two groups. The group receiving therapy included patients treated with prednisolone (PSL), mycophenolate mofetil (MMF), CsA, methotrexate (MTX), anti-TNF therapy, or a combination. A higher CD8+ TSCM frequency (4.8%) was observed in uveitis patients without any systemic immune therapy compared with those receiving immune therapies (3.2%) (p < 0.05, Fig. 6A). A higher CD8+ TSCM frequency was observed in uveitis patients not receiving PSL (4.1%) than in those receiving PSL (2.8%), as well as in those patients not receiving the anti-TNF agents infliximab and adalimumab (4.0%) than in patients receiving these drugs (2.4%) (p < 0.05, Fig. 6A). However, there were no statistically significant differences in patients with or without CsA, MTX, or MMF (p > 0.05, Supplemental Fig. 3B). CD8+ TSCM frequency did not correlate with age, sex, diagnosis of underlying disease (sarcoidosis versus Vogt–Koyanagi–Harada disease versus Birdshot), localization of disease, or disease activity (p > 0.05) (Supplemental Fig. 3B).

FIGURE 6.

Clinical correlations with the TSCM populations in uveitis and SLE patients. (A) Frequency of CD8+ TSCMs in uveitis (n = 34) compared between the following groups: with and without immune therapies (including one drug alone or any combination of drugs), with and without PSL (PSL alone or PSL plus any other drugs), and with and without anti-TNF (anti-TNF alone or anti-TNF plus any other drugs). (B) CD4+ TSCM frequency in SLE (n = 43) was compared between patients with and without HCQ (HCQ alone or HCQ plus any other drugs). *p < 0.05.

FIGURE 6.

Clinical correlations with the TSCM populations in uveitis and SLE patients. (A) Frequency of CD8+ TSCMs in uveitis (n = 34) compared between the following groups: with and without immune therapies (including one drug alone or any combination of drugs), with and without PSL (PSL alone or PSL plus any other drugs), and with and without anti-TNF (anti-TNF alone or anti-TNF plus any other drugs). (B) CD4+ TSCM frequency in SLE (n = 43) was compared between patients with and without HCQ (HCQ alone or HCQ plus any other drugs). *p < 0.05.

Close modal

Because most SLE patients had received IST before or at the time of sampling, we could not examine therapy effects on the TSCM population. There was a higher frequency of CD4+ TSCMs in patients not receiving hydroxychloroquine (HCQ; 3.3%) than in those receiving HCQ (2.3%) (p < 0.05, Fig. 6B), but no relationship was seen for any other drug (Supplemental Fig. 3C). CD4+ TSCM frequency was not correlated with age, disease activity (p > 0.05; Supplemental Fig. 3C), or the presence of specific disease associations (including secondary Sjögren’s syndrome, antiphospholipid syndrome, or immune thrombocytopenic purpura) (data not shown).

Collectively, immune therapies appeared to have negative effects on the TSCM population in the uveitis and SLE patient cohorts, as well as in AA.

In our flow cytometry–based study, we retrospectively analyzed CD4+ and CD8+ TSCM populations and their relationship to disease activity, treatment response, and disease severity by characterizing these subsets in a total of 178 subjects (55 AA, 34 autoimmune uveitis, 43 SLE, 5 SCD, and 41 age-matched healthy controls). An increased CD8+ TSCM population was observed in AA, uveitis, and SCD, and a higher tendency of CD4+ TSCM population was seen in SLE. Activation of CD4+ and CD8+ T cells is critical in diseases of uncontrolled inflammation (5, 28, 29), but we believe the current study to be the first report of CD4+ and CD8+ TSCM populations in autoimmune diseases. Consistent with previous studies, we observed ∼2–4% TSCMs in the total CD4+ or CD8+ T cell population in healthy controls (12, 18), and there was no correlation between CD4+ or CD8+ TSCM frequency and age in any group (30).

TSCMs have been examined in infectious diseases (25, 31, 32), in the development of immunotherapies and vaccines for cancer (33, 34), in allogeneic hematopoietic stem cell transplantation (35, 36), and in adult T cell leukemia (37). TSCMs represent the earliest and longest-lasting developmental stage of memory T cells and exhibit a gene expression profile between TNs and TCMs (12, 18). One mechanism for the maintenance of long-term T cell memory may be the unique homeostatic properties of TSCMs (38). In this context, we observed an inverse correlation between the CD4+ or CD8+ TSCM population and the CD4+ TEM or CD8+ TE population, respectively, indicating that TSCM differentiation leads to a shift in these cell proportions as a possible mechanism to re-establish the homeostasis of the immune system. TSCMs may be progenitors for TEMs or TEs, because these two proportions were inversely correlated. It is unknown whether immune activation is attributable to increased TSCMs or the increase in this subset is the result of aberrant immune responses. Because of their capacity to generate all memory and effector T cell subsets, we hypothesized that the increased frequency of TSCMs contributed to the progression of autoimmune diseases.

Indeed, a higher CD8+ TSCM frequency at diagnosis or after IST was associated with a better response to IST or treatment failure, respectively, in AA, suggesting CD8+ TSCMs as a potential biomarker. Longitudinal analysis of 13 AA patients also suggested a potential use for TSCMs as a disease biomarker. In the uveitis cohort, the CD8+ TSCM population was lower in patients who received PSL and anti-TNF therapy, indicating a potential negative role for IST in this population, although the analysis of serial samples would be required to support this conclusion. Further, SLE patients receiving HCQ therapy displayed a reduced CD4+ TSCM frequency, also suggesting that IST affected this population. HCQ is known to correlate negatively with the percentage of CD45RO+ cells among CD4+ T cells (39). Because most SLE samples were collected after IST, IST might have masked the TSCM population. Additional studies in large patient cohorts are necessary to define whether TSCMs are useful as general autoimmune disease biomarkers.

There are similarities and differences in T cell autoimmunity in AA, uveitis, and SLE. Abnormalities of regulatory T cells and Th17 cells are reported in patients with AA (4042), uveitis (43, 44), or SLE (4547). CD8+ CTLs with restricted TCR diversity are expanded in AA and secrete proinflammatory cytokines, which induce apoptosis of CD34+ cells (6). A recent study suggested that the clonally restricted expansion of Th1 cells is most likely Ag-driven and induces an inflammatory environment, exacerbating the functional impairment of regulatory T cells (40). In uveitis, a CD4+ T cell–driven disease is the dominant paradigm in animal models (28). However, Ag-specific CD8+ T cells can mediate autoimmunity within the eye (48), and CD8+ T cells increase in experimental uveitis (49). SLE is characterized by the production of a wide array of autoantibodies and, thus, has traditionally been classified as a B cell disease. However, evidence indicated that the assistance of Th cells might be required to produce SLE-related inflammation (50).

Upon TCR stimulation in healthy controls, TSCMs exhibit effector activity, including TNF-α, IFN-γ, and IL-2 secretion, whereas TNs remain relatively quiescent (12). In AA patients, CD4+ and CD8+ TSCMs produced more IFN-γ and IL-2 upon TCR stimulation compared with healthy controls, showing the effector functions and greater IL-2–producing capacity of these subsets. CD8+ CTLs are expanded in AA and produce proinflammatory cytokines, such as IFN-γ, which induce apoptosis of CD34+ cells (5). In this study, we identified TSCMs as another source of IFN-γ production by CD4+ and CD8+ T cells in AA.

PD-1 is one of the immune checkpoint molecules expressed on activated and exhausted T cells (51). PD-1 overexpression is detected in CD4+ and CD8+ T cells in AA patients (21). It was demonstrated in SLE patients that PD-1 expression is upregulated in CD4+ T cells, and that PD-1 expression in CD4+ T cells is associated with IFN-γ expression in CD3+ T cells (52). In this study, PD-1 expression in TSCMs identified this subset as the least exhausted population relative to other memory T cell subsets. Previous studies demonstrated PD-1 expression on tumor-infiltrating T cells (TILs) and its correlation with prognosis (53, 54). The PD-1 pathway exerts inhibitory functions in chronic viral infections and tumors, with special relevance to autoimmunity (23). In addition to well-documented negative-regulatory roles, PD-1 expression on CD8+ TILs accurately identified the repertoire of clonally expanded tumor-reactive cells (55). The higher expression of PD-1 in CD8+ TSCMs in AA suggests this population as self-reactive T cells. Recent studies suggested that PD-1 expression in healthy donors does not correlate with lower functionality but rather with differentiation to an effector memory phenotype (19, 26, 56). We interpret increased expression of PD-1 and increased IFN-γ production in CD8+ TSCMs of AA patients as evidence of clonal expansion, as occurs for CD8+ TILs (55) and as we reported previously for CD8+ effector cells in AA (6, 57). Enhanced cytolytic effector activity in CD8+ TSCMs of AA was also supported by the observation of higher CD160 expression of this population.

Our study has a number of limitations. We were unable to demonstrate causality. However, it seems likely that an increase in TSCMs may be etiologically associated with immune response to autoantigens and that the mechanism may be related to homeostatic maintenance of other memory T cell subsets. It is also plausible that increased TSCMs are a consequence of nonspecific inflammation induced by environmental factors, because CD8+ TSCMs were also increased in SCD, which is not typically considered an autoimmune disease. Transfusion is chronic immune stimulation, which may drive SCD to an active inflammatory state (58, 59). Transfusions administered to AA and SCD patients may affect the CD8+ TSCM population as a result of allogeneic stimulation. However, this seems unlikely because CD8+ TSCM frequency did not correlate with serum ferritin levels in AA (Supplemental Fig. 3A). Although we were unable to characterize the Ag-specific TSCM population because of the limited cell numbers, a nonhuman primate model of SIV infection demonstrated that SIV-specific TSCMs preferentially survive after Ag elimination compared with other memory subsets and are fully functional, even in chronic infection (38). Thus, Ag-specific TSCMs are presumably able to contribute directly to disease progression.

In conclusion, we provide evidence for increased circulating CD8+ TSCMs in AA, underscoring the importance of this novel subset in the regulation of immune responses and pathogenesis of autoimmunity. Our work described previously unknown potential roles for TSCMs in AA, such as cytokine secretion correlated with effector functions. Understanding the CD8+ TSCM population may offer new therapeutic strategies and novel mechanistic insights into various autoimmune diseases. Longitudinal analysis of AA patients suggests the potential use of TSCMs as disease biomarkers. Additional studies in large patient cohorts are required to validate our data, which were obtained from small numbers of patients with autoimmune diseases.

We thank Marie Desierto, Susan Wong, and Pilar Fernandez for technical assistance; Olga Rios, Kinneret Broder, and Carolyne Smith for assistance with obtaining patient and healthy volunteer samples; and Barbara Weinstein, Sarfaraz Hasni, and Zerai Manna for obtaining patient clinical information.

This work was supported by the Intramural Research Program of the National Institutes of Health, National Heart, Lung, and Blood Institute.

The online version of this article contains supplemental material.

Abbreviations used in this article:

AA

aplastic anemia

ATG

anti-thymocyte globulin

BM

bone marrow

CR

complete response

CsA

cyclosporine A

GZMB

granzyme B

HCQ

hydroxychloroquine

HSPC

hematopoietic stem/progenitor cell

IST

immunosuppressive therapy

MMF

mycophenolate mofetil

MTX

methotrexate

NR

nonresponder

PB

peripheral blood

PR

partial response

PSL

prednisolone

SCD

sickle cell disease

SLE

systemic lupus erythematosus

SLEDAI-2K

SLE disease activity index-2K

TCM

central memory T cell

TE

terminally differentiated TEM

TEM

effector memory T cell

TIL

tumor-infiltrating cell

TN

naive T cell

TSCM

memory stem T cell.

1
Young
N. S.
,
Calado
R. T.
,
Scheinberg
P.
.
2006
.
Current concepts in the pathophysiology and treatment of aplastic anemia.
Blood
108
:
2509
2519
.
2
Scheinberg
P.
,
Young
N. S.
.
2012
.
How I treat acquired aplastic anemia.
Blood
120
:
1185
1196
.
3
Risitano
A. M.
,
Kook
H.
,
Zeng
W.
,
Chen
G.
,
Young
N. S.
,
Maciejewski
J. P.
.
2002
.
Oligoclonal and polyclonal CD4 and CD8 lymphocytes in aplastic anemia and paroxysmal nocturnal hemoglobinuria measured by V beta CDR3 spectratyping and flow cytometry.
Blood
100
:
178
183
.
4
Nakao
S.
,
Takami
A.
,
Takamatsu
H.
,
Zeng
W.
,
Sugimori
N.
,
Yamazaki
H.
,
Miura
Y.
,
Ueda
M.
,
Shiobara
S.
,
Yoshioka
T.
, et al
.
1997
.
Isolation of a T-cell clone showing HLA-DRB1*0405-restricted cytotoxicity for hematopoietic cells in a patient with aplastic anemia.
Blood
89
:
3691
3699
.
5
Sloand
E.
,
Kim
S.
,
Maciejewski
J. P.
,
Tisdale
J.
,
Follmann
D.
,
Young
N. S.
.
2002
.
Intracellular interferon-gamma in circulating and marrow T cells detected by flow cytometry and the response to immunosuppressive therapy in patients with aplastic anemia.
Blood
100
:
1185
1191
.
6
Risitano
A. M.
,
Maciejewski
J. P.
,
Green
S.
,
Plasilova
M.
,
Zeng
W.
,
Young
N. S.
.
2004
.
In-vivo dominant immune responses in aplastic anaemia: molecular tracking of putatively pathogenetic T-cell clones by TCR beta-CDR3 sequencing.
Lancet
364
:
355
364
.
7
Maciejewski
J. P.
,
Follmann
D.
,
Nakamura
R.
,
Saunthararajah
Y.
,
Rivera
C. E.
,
Simonis
T.
,
Brown
K. E.
,
Barrett
J. A.
,
Young
N. S.
.
2001
.
Increased frequency of HLA-DR2 in patients with paroxysmal nocturnal hemoglobinuria and the PNH/aplastic anemia syndrome.
Blood
98
:
3513
3519
.
8
Katagiri
T.
,
Sato-Otsubo
A.
,
Kashiwase
K.
,
Morishima
S.
,
Sato
Y.
,
Mori
Y.
,
Kato
M.
,
Sanada
M.
,
Morishima
Y.
,
Hosokawa
K.
, et al
Japan Marrow Donor Program
.
2011
.
Frequent loss of HLA alleles associated with copy number-neutral 6pLOH in acquired aplastic anemia.
Blood
118
:
6601
6609
.
9
Scheinberg
P.
,
Nunez
O.
,
Weinstein
B.
,
Scheinberg
P.
,
Biancotto
A.
,
Wu
C. O.
,
Young
N. S.
.
2011
.
Horse versus rabbit antithymocyte globulin in acquired aplastic anemia.
N. Engl. J. Med.
365
:
430
438
.
10
Sallusto
F.
,
Lenig
D.
,
Förster
R.
,
Lipp
M.
,
Lanzavecchia
A.
.
1999
.
Two subsets of memory T lymphocytes with distinct homing potentials and effector functions.
Nature
401
:
708
712
.
11
Hu
X.
,
Gu
Y.
,
Wang
Y.
,
Cong
Y.
,
Qu
X.
,
Xu
C.
.
2009
.
Increased CD4+ and CD8+ effector memory T cells in patients with aplastic anemia.
Haematologica
94
:
428
429
.
12
Gattinoni
L.
,
Lugli
E.
,
Ji
Y.
,
Pos
Z.
,
Paulos
C. M.
,
Quigley
M. F.
,
Almeida
J. R.
,
Gostick
E.
,
Yu
Z.
,
Carpenito
C.
, et al
.
2011
.
A human memory T cell subset with stem cell-like properties.
Nat. Med.
17
:
1290
1297
.
13
Gattinoni
L.
2014
.
Memory T cells officially join the stem cell club.
Immunity
41
:
7
9
.
14
1987
.
Incidence of aplastic anemia: the relevance of diagnostic criteria. By the International Agranulocytosis and Aplastic Anemia Study.
Blood
70
:
1718
1721
.
15
Camitta
B. M.
,
Thomas
E. D.
,
Nathan
D. G.
,
Santos
G.
,
Gordon-Smith
E. C.
,
Gale
R. P.
,
Rappeport
J. M.
,
Storb
R.
.
1976
.
Severe aplastic anemia: a prospective study of the effect of early marrow transplantation on acute mortality.
Blood
48
:
63
70
.
16
Hochberg
M. C.
1997
.
Updating the American College of Rheumatology revised criteria for the classification of systemic lupus erythematosus.
Arthritis Rheum.
40
:
1725
.
17
Gladman
D. D.
,
Ibañez
D.
,
Urowitz
M. B.
.
2002
.
Systemic lupus erythematosus disease activity index 2000.
J. Rheumatol.
29
:
288
291
.
18
Lugli
E.
,
Gattinoni
L.
,
Roberto
A.
,
Mavilio
D.
,
Price
D. A.
,
Restifo
N. P.
,
Roederer
M.
.
2013
.
Identification, isolation and in vitro expansion of human and nonhuman primate T stem cell memory cells.
Nat. Protoc.
8
:
33
42
.
19
Legat
A.
,
Speiser
D. E.
,
Pircher
H.
,
Zehn
D.
,
Fuertes Marraco
S. A.
.
2013
.
Inhibitory Receptor Expression Depends More Dominantly on Differentiation and Activation than “Exhaustion” of Human CD8 T Cells.
Front. Immunol.
4
:
455
.
20
Camitta
B. M.
,
Doney
K.
.
1990
.
Immunosuppressive therapy for aplastic anemia: indications, agents, mechanisms, and results.
Am. J. Pediatr. Hematol. Oncol.
12
:
411
424
.
21
Wu
H.
,
Miao
M.
,
Zhang
G.
,
Hu
Y.
,
Ming
Z.
,
Zhang
X.
.
2009
.
Soluble PD-1 is associated with aberrant regulation of T cells activation in aplastic anemia.
Immunol. Invest.
38
:
408
421
.
22
Jiao
Q.
,
Liu
C.
,
Yang
Z.
,
Ding
Q.
,
Wang
M.
,
Li
M.
,
Zhu
T.
,
Qian
H.
,
Li
W.
,
Tu
N.
, et al
.
2014
.
Upregulated PD-1 Expression Is Associated with the Development of Systemic Lupus Erythematosus, but Not the PD-1.1 Allele of the PDCD1 Gene.
Int. J. Genomics
2014
:
950903
.
23
Gianchecchi
E.
,
Delfino
D. V.
,
Fierabracci
A.
.
2013
.
Recent insights into the role of the PD-1/PD-L1 pathway in immunological tolerance and autoimmunity.
Autoimmun. Rev.
12
:
1091
1100
.
24
Adekambi
T.
,
Ibegbu
C. C.
,
Kalokhe
A. S.
,
Yu
T.
,
Ray
S. M.
,
Rengarajan
J.
.
2012
.
Distinct effector memory CD4+ T cell signatures in latent Mycobacterium tuberculosis infection, BCG vaccination and clinically resolved tuberculosis.
PLoS One
7
:
e36046
.
25
Jaafoura
S.
,
de Goër de Herve
M. G.
,
Hernandez-Vargas
E. A.
,
Hendel-Chavez
H.
,
Abdoh
M.
,
Mateo
M. C.
,
Krzysiek
R.
,
Merad
M.
,
Seng
R.
,
Tardieu
M.
, et al
.
2014
.
Progressive contraction of the latent HIV reservoir around a core of less-differentiated CD4⁺ memory T Cells.
Nat. Commun.
5
:
5407
.
26
Baitsch
L.
,
Legat
A.
,
Barba
L.
,
Fuertes Marraco
S. A.
,
Rivals
J. P.
,
Baumgaertner
P.
,
Christiansen-Jucht
C.
,
Bouzourene
H.
,
Rimoldi
D.
,
Pircher
H.
, et al
.
2012
.
Extended co-expression of inhibitory receptors by human CD8 T-cells depending on differentiation, antigen-specificity and anatomical localization.
PLoS One
7
:
e30852
.
27
Riches
J. C.
,
Davies
J. K.
,
McClanahan
F.
,
Fatah
R.
,
Iqbal
S.
,
Agrawal
S.
,
Ramsay
A. G.
,
Gribben
J. G.
.
2013
.
T cells from CLL patients exhibit features of T-cell exhaustion but retain capacity for cytokine production.
Blood
121
:
1612
1621
.
28
Lee
R. W.
,
Nicholson
L. B.
,
Sen
H. N.
,
Chan
C. C.
,
Wei
L.
,
Nussenblatt
R. B.
,
Dick
A. D.
.
2014
.
Autoimmune and autoinflammatory mechanisms in uveitis.
Semin. Immunopathol.
36
:
581
594
.
29
Gualtierotti
R.
,
Biggioggero
M.
,
Penatti
A. E.
,
Meroni
P. L.
.
2010
.
Updating on the pathogenesis of systemic lupus erythematosus.
Autoimmun. Rev.
10
:
3
7
.
30
Van Epps
P.
,
Banks
R.
,
Aung
H.
,
Betts
M. R.
,
Canaday
D. H.
.
2014
.
Age-related differences in polyfunctional T cell responses.
Immun. Ageing
11
:
14
.
31
Mateus
J.
,
Lasso
P.
,
Pavia
P.
,
Rosas
F.
,
Roa
N.
,
Valencia-Hernández
C. A.
,
González
J. M.
,
Puerta
C. J.
,
Cuéllar
A.
.
2015
.
Low frequency of circulating CD8+ T stem cell memory cells in chronic chagasic patients with severe forms of the disease.
PLoS Negl. Trop. Dis.
9
:
e3432
.
32
Klatt
N. R.
,
Bosinger
S. E.
,
Peck
M.
,
Richert-Spuhler
L. E.
,
Heigele
A.
,
Gile
J. P.
,
Patel
N.
,
Taaffe
J.
,
Julg
B.
,
Camerini
D.
, et al
.
2014
.
Limited HIV infection of central memory and stem cell memory CD4+ T cells is associated with lack of progression in viremic individuals.
PLoS Pathog.
10
:
e1004345
.
33
Gattinoni
L.
,
Restifo
N. P.
.
2013
.
Moving T memory stem cells to the clinic.
Blood
121
:
567
568
.
34
Flynn
J. K.
,
Gorry
P. R.
.
2014
.
Stem memory T cells (TSCM)-their role in cancer and HIV immunotherapies.
Clin. Transl. Immunology
3
:
e20
.
35
Cieri
N.
,
Oliveira
G.
,
Greco
R.
,
Forcato
M.
,
Taccioli
C.
,
Cianciotti
B.
,
Valtolina
V.
,
Noviello
M.
,
Vago
L.
,
Bondanza
A.
, et al
.
2015
.
Generation of human memory stem T cells after haploidentical T-replete hematopoietic stem cell transplantation.
Blood
125
:
2865
2874
.
36
Roberto
A.
,
Castagna
L.
,
Zanon
V.
,
Bramanti
S.
,
Crocchiolo
R.
,
McLaren
J. E.
,
Gandolfi
S.
,
Tentorio
P.
,
Sarina
B.
,
Timofeeva
I.
, et al
.
2015
.
Role of naive-derived T memory stem cells in T-cell reconstitution following allogeneic transplantation.
Blood
125
:
2855
2864
.
37
Nagai
Y.
,
Kawahara
M.
,
Hishizawa
M.
,
Shimazu
Y.
,
Sugino
N.
,
Fujii
S.
,
Kadowaki
N.
,
Takaori-Kondo
A.
.
2015
.
T memory stem cells are the hierarchical apex of adult T-cell leukemia.
Blood
125
:
3527
3535
.
38
Lugli
E.
,
Dominguez
M. H.
,
Gattinoni
L.
,
Chattopadhyay
P. K.
,
Bolton
D. L.
,
Song
K.
,
Klatt
N. R.
,
Brenchley
J. M.
,
Vaccari
M.
,
Gostick
E.
, et al
.
2013
.
Superior T memory stem cell persistence supports long-lived T cell memory.
J. Clin. Invest.
123
:
594
599
.
39
Sailler
L.
,
Puissant
B.
,
Méliani
P.
,
Castex
J. O.
,
Saivin
S.
,
Adoue
D.
,
Fournie
B.
,
Arlet
P.
,
Montastruc
J. L.
,
Lapeyre-Mestre
M.
, et al
.
2007
.
Blood concentrations of hydroxychloroquine and its desethyl derivative correlate negatively with the percentage of CD45RO+ cells among CD4+ lymphocytes in hydroxychloroquine-treated lupus patients.
Ann. N. Y. Acad. Sci.
1108
:
41
50
.
40
Kordasti
S.
,
Marsh
J.
,
Al-Khan
S.
,
Jiang
J.
,
Smith
A.
,
Mohamedali
A.
,
Abellan
P. P.
,
Veen
C.
,
Costantini
B.
,
Kulasekararaj
A. G.
, et al
.
2012
.
Functional characterization of CD4+ T cells in aplastic anemia.
Blood
119
:
2033
2043
.
41
Solomou
E. E.
,
Rezvani
K.
,
Mielke
S.
,
Malide
D.
,
Keyvanfar
K.
,
Visconte
V.
,
Kajigaya
S.
,
Barrett
A. J.
,
Young
N. S.
.
2007
.
Deficient CD4+ CD25+ FOXP3+ T regulatory cells in acquired aplastic anemia.
Blood
110
:
1603
1606
.
42
de Latour
R. P.
,
Visconte
V.
,
Takaku
T.
,
Wu
C.
,
Erie
A. J.
,
Sarcon
A. K.
,
Desierto
M. J.
,
Scheinberg
P.
,
Keyvanfar
K.
,
Nunez
O.
, et al
.
2010
.
Th17 immune responses contribute to the pathophysiology of aplastic anemia.
Blood
116
:
4175
4184
.
43
Yeh
S.
,
Li
Z.
,
Forooghian
F.
,
Hwang
F. S.
,
Cunningham
M. A.
,
Pantanelli
S.
,
Lew
J. C.
,
Wroblewski
K. K.
,
Vitale
S.
,
Nussenblatt
R. B.
.
2009
.
CD4+Foxp3+ T-regulatory cells in noninfectious uveitis.
Arch. Ophthalmol.
127
:
407
413
.
44
Amadi-Obi
A.
,
Yu
C. R.
,
Liu
X.
,
Mahdi
R. M.
,
Clarke
G. L.
,
Nussenblatt
R. B.
,
Gery
I.
,
Lee
Y. S.
,
Egwuagu
C. E.
.
2007
.
TH17 cells contribute to uveitis and scleritis and are expanded by IL-2 and inhibited by IL-27/STAT1.
Nat. Med.
13
:
711
718
.
45
Martin
J. C.
,
Baeten
D. L.
,
Josien
R.
.
2014
.
Emerging role of IL-17 and Th17 cells in systemic lupus erythematosus.
Clin. Immunol.
154
:
1
12
.
46
Mellor-Pita
S.
,
Citores
M. J.
,
Castejon
R.
,
Tutor-Ureta
P.
,
Yebra-Bango
M.
,
Andreu
J. L.
,
Vargas
J. A.
.
2006
.
Decrease of regulatory T cells in patients with systemic lupus erythematosus.
Ann. Rheum. Dis.
65
:
553
554
.
47
Shin
M. S.
,
Lee
N.
,
Kang
I.
.
2011
.
Effector T-cell subsets in systemic lupus erythematosus: update focusing on Th17 cells.
Curr. Opin. Rheumatol.
23
:
444
448
.
48
McPherson
S. W.
,
Yang
J.
,
Chan
C. C.
,
Dou
C.
,
Gregerson
D. S.
.
2003
.
Resting CD8 T cells recognize beta-galactosidase expressed in the immune-privileged retina and mediate autoimmune disease when activated.
Immunology
110
:
386
396
.
49
Calder
V. L.
,
Zhao
Z. S.
,
Wang
Y.
,
Barton
K.
,
Lightman
S. L.
.
1993
.
Effects of CD8 depletion on retinal soluble antigen induced experimental autoimmune uveoretinitis.
Immunology
79
:
255
262
.
50
Mak
A.
,
Kow
N. Y.
.
2014
.
The pathology of T cells in systemic lupus erythematosus.
J. Immunol. Res.
2014
:
419029
.
51
Kamphorst
A. O.
,
Ahmed
R.
.
2013
.
Manipulating the PD-1 pathway to improve immunity.
Curr. Opin. Immunol.
25
:
381
388
.
52
Dolff
S.
,
Quandt
D.
,
Feldkamp
T.
,
Jun
C.
,
Mitchell
A.
,
Hua
F.
,
Specker
C.
,
Kribben
A.
,
Witzke
O.
,
Wilde
B.
.
2014
.
Increased percentages of PD-1 on CD4+ T cells is associated with higher INF-γ production and altered IL-17 production in patients with systemic lupus erythematosus.
Scand. J. Rheumatol.
43
:
307
313
.
53
Thompson
R. H.
,
Dong
H.
,
Lohse
C. M.
,
Leibovich
B. C.
,
Blute
M. L.
,
Cheville
J. C.
,
Kwon
E. D.
.
2007
.
PD-1 is expressed by tumor-infiltrating immune cells and is associated with poor outcome for patients with renal cell carcinoma.
Clin. Cancer Res.
13
:
1757
1761
.
54
Konishi
J.
,
Yamazaki
K.
,
Azuma
M.
,
Kinoshita
I.
,
Dosaka-Akita
H.
,
Nishimura
M.
.
2004
.
B7-H1 expression on non-small cell lung cancer cells and its relationship with tumor-infiltrating lymphocytes and their PD-1 expression.
Clin. Cancer Res.
10
:
5094
5100
.
55
Gros
A.
,
Robbins
P. F.
,
Yao
X.
,
Li
Y. F.
,
Turcotte
S.
,
Tran
E.
,
Wunderlich
J. R.
,
Mixon
A.
,
Farid
S.
,
Dudley
M. E.
, et al
.
2014
.
PD-1 identifies the patient-specific CD8+ tumor-reactive repertoire infiltrating human tumors.
J. Clin. Invest.
124
:
2246
2259
.
56
Duraiswamy
J.
,
Ibegbu
C. C.
,
Masopust
D.
,
Miller
J. D.
,
Araki
K.
,
Doho
G. H.
,
Tata
P.
,
Gupta
S.
,
Zilliox
M. J.
,
Nakaya
H. I.
, et al
.
2011
.
Phenotype, function, and gene expression profiles of programmed death-1(hi) CD8 T cells in healthy human adults.
J. Immunol.
186
:
4200
4212
.
57
Kook
H.
,
Risitano
A. M.
,
Zeng
W.
,
Wlodarski
M.
,
Lottemann
C.
,
Nakamura
R.
,
Barrett
J.
,
Young
N. S.
,
Maciejewski
J. P.
.
2002
.
Changes in T-cell receptor VB repertoire in aplastic anemia: effects of different immunosuppressive regimens.
Blood
99
:
3668
3675
.
58
Fasano
R. M.
,
Booth
G. S.
,
Miles
M.
,
Du
L.
,
Koyama
T.
,
Meier
E. R.
,
Luban
N. L.
.
2015
.
Red blood cell alloimmunization is influenced by recipient inflammatory state at time of transfusion in patients with sickle cell disease.
Br. J. Haematol.
168
:
291
300
.
59
Lee
S. P.
,
Ataga
K. I.
,
Orringer
E. P.
,
Phillips
D. R.
,
Parise
L. V.
.
2006
.
Biologically active CD40 ligand is elevated in sickle cell anemia: potential role for platelet-mediated inflammation.
Arterioscler. Thromb. Vasc. Biol.
26
:
1626
1631
.

The authors have no financial conflicts of interest.

Supplementary data