Senescent T cells have been implicated in chronic inflammatory and cardiovascular diseases. In this study, we explored the relationship between senescent T cells and glycemic status in a cohort of 805 participants by investigating the frequency of CD57+ or CD28null senescent T cells in peripheral blood. Participants with normal glucose tolerance (NGT) with follow-up data (N = 149) were included to determine whether hyperglycemia (prediabetes or type 2 diabetes) developed during follow-up (mean 2.3 years). CD8+CD57+ and CD8+CD28null T-cell frequencies were significantly higher in prediabetes and type 2 diabetes compared with NGT. Increased CD57+ or CD28null cells in the CD8+ T-cell subset were independently associated with hyperglycemia. Furthermore, among participants with baseline NGT, the frequency of CD8+CD57+ T cells was an independent predictor of hyperglycemia development. Immunofluorescent analyses confirmed that CD8+CD57+ T-cell infiltration was increased in visceral adipose tissue of patients with prediabetes or type 2 diabetes compared with those with NGT. Our data suggest that increased frequency of senescent CD8+ T cells in the peripheral blood is associated with development of hyperglycemia.

Immunosenescence is the progressive impairment of the immune system thought to underlie age-related comorbidities (1,2). Change in T-cell immunities is a notable feature of immunosenescence. There is accumulating evidence that senescent T cells are involved in the pathogenesis of cardiovascular diseases (CVDs), including atherosclerosis, acute coronary syndrome, and hypertension (37). We reported that hypertensive patients had an increased frequency of senescent CD8+ T cells in peripheral blood, which exhibited CD28 loss and acquisition of CD57 on their surface (8). CD28 loss is a prominent change associated with human aging and is caused by the repetitive antigenic stimulation of T cells. CD57 expression during the late stage of T-cell differentiation might be a distinct measure of senescence in T cells (9). Compared with CD28+ or CD57null T cells, CD28null or CD57+ T cells produce more proinflammatory cytokines and exert greater cytotoxicity (10).

Substantial overlap and interconnectivity exists between the etiology and pathophysiology of diabetes and CVD, theorized as the “common soil” hypothesis (11). Despite evidence that immunosenescence can lead to CVD, there are no studies evaluating whether increased T-cell senescence leads to hyperglycemia development in humans. Our study’s aim was to investigate the relationship between senescent T cells and glycemic status using a cross-sectional cohort. Furthermore, we studied the longitudinal impact of senescent T cells on hyperglycemia development in participants with normoglycemia.

Study Participants

In this prospective longitudinal study, 805 Koreans registered in the Yonsei Cardiovascular Genome cohort or the Cardiovascular and Metabolic Disease Etiology Research Center-High risk cohort (clinicaltrials.gov: NCT02003781) were recruited via the outpatient clinic of Severance Cardiovascular Hospital from January 2011 to April 2016. Type 2 diabetes was defined as: fasting plasma glucose level ≥126 mg/dL; hemoglobin A1c (HbA1c) ≥6.5% (48 mmol/mol); or history of insulin or oral hypoglycemic agent administration. Participants without diabetes were categorized as either normal glucose tolerance (NGT), defined as fasting plasma glucose levels <100 mg/dL and HbA1c <5.7% (39 mmol/mol) or prediabetes, defined as fasting plasma glucose levels 100–125 mg/dL or HbA1c 5.7–6.4% (39–46 mmol/mol) (12). Hyperglycemia included prediabetes and type 2 diabetes (13). Hypertension was defined as systolic blood pressure >140 mmHg or diastolic blood pressure >90 mmHg over three visits before the use of antihypertensives. Coronary artery disease was defined as one or more lesions with a >50% diameter reduction by coronary angiography. Patients with any of the following conditions were excluded: significant systemic disease, debilitating malignant disease, severe hypertension (>200/140 mmHg), estimated glomerular filtration rate <30 mL/min/1.73 m2, and history of overt chronic inflammatory disease and/or receiving anti-inflammatory medications. This study was approved by the Yonsei University College of Medicine Institutional Review Board, and study procedures were in accordance with institutional guidelines (Institutional Review Board number 4-2018-0190). Participants provided informed consent before enrollment.

Blood Glucometabolic Parameters and Incidence of Hyperglycemia

Following an overnight fast, blood tests for biochemical measurements were performed. Serum glucose, insulin, and HbA1c were measured using the Hitachi 7600 analyzer (Hitachi Ltd.), immunoradiometric assay (Insulin-IRMA; DIAsource, Louvain-la-Neuve, Belgium), and immunoassay by an Integra 800 CTS (Roche Diagnostics), respectively. We examined medical records of participants with normoglycemia who had at least one clinic visit for any reason from the date of registry enrollment until December 2016. Of 222 NGT participants, follow-up blood glucose data were available in 149 (88 men and 61 women). Based on fasting plasma glucose or HbA1c levels, participants with baseline NGT were defined as hyperglycemia progressors when those met diagnostic criteria for prediabetes or diabetes in at least one time interval.

Immunophenotype Analysis of Peripheral Blood Mononuclear Cells

Peripheral blood mononuclear cells (PBMCs) were isolated using Ficoll-Hypaque (GE Healthcare, Uppsala, Sweden) density gradient centrifugation and immediately stained for flow cytometry analyses. PBMCs were incubated with directly conjugated monoclonal antibodies for 20 min at 4°C using anti-CD3 (Horizon V500), anti-CD4 (phycoerythrin [PE]-Cy7), anti-CD8 (allophycocyanin [APC]-H7), anti-CD19 (PerCP-Cy5.5), anti-CD28 (APC) (all from BD Biosciences, San Jose, CA), and anti-CD57 (eFluor 450; BioLegend, San Diego, CA). Multicolor flow cytometry was performed using an LSR II instrument (BD Biosciences) and analyzed using FlowJo software (Tree Star, San Carlos, CA). The gating strategy is provided in Supplementary Fig. 1. FlowJo software autogating was performed on CD28 and CD57 T cells. Frequency of CD8+CD57+ or CD8+CD28null T cells and CD4+CD57+ or CD4+CD28null T cells was expressed as a percentage of the entire population of CD8+ and CD4+ T cells.

In Vitro Stimulation of T Cells and Intracellular Cytokine Staining

Cytomegalovirus (CMV) serostatus was evaluated using a chemiluminescent microparticle immunoassay (Abbott Laboratories, Chicago, IL), and a titer of ≥6.0 antibody units/mL of IgG was considered CMV IgG-seropositive. PBMCs were stimulated with overlapping peptides from CMV pp65 (0.6 nmol of each peptide/mL) (Miltenyi Biotec) for 6 h in the presence of PE-conjugated anti-CD107a (BD Biosciences). After 1 h of incubation, brefeldin A (GolgiPlug; BD Biosciences) and monensin (GolgiStop; BD Biosciences) were added to accumulate cytokine proteins intracellularly. Following surface staining with anti-CD3 (Horizon V500), anti-CD4 (PerCP-Cy5.5), anti-CD8 (APC-H7), anti-CD28 (Horizon V450), and anti-CD57 (APC), the cells were fixed and permeabilized using a Fixation/Permeabilization Buffer Kit and further stained for intracellular cytokines with anti–interferon-γ (FITC) and anti–tumor necrosis factor-α (PE-Cy7) (both from BD Biosciences). All samples were assessed using an LSR II Flow Cytometer (BD Biosciences), and the data were analyzed using FlowJo software.

Adipose Tissue Immunohistochemistry and Immunofluorescence Staining

Omental adipose tissue from consenting patients without acute infection or receiving immune-modulating medications was obtained during abdominal surgery (e.g., hepatectomy, colectomy, or cholecystectomy due to cancer). In order to identify infiltrated CD8+ T cells adjacent to macrophages within adipose tissues, immunohistochemistry was performed with formalin-fixed, paraffin-embedded sections using the primary antibodies of CD68 (Thermo Fisher Scientific, Waltham, MA) and CD8 (Abcam, Cambridge, U.K.) and the secondary antibody of Polink DS (GBI Labs, Bothell, WA). Next, double immunofluorescence staining was performed to colocalize CD57+ with CD8+ T cells. Primary antibodies used were CD57 (HNK-1, Leu-7, and MA5-11605; Thermo Fisher Scientific) and CD8 (ab4055; Abcam). Secondary antibodies used were Alexa Fluor 488 (A21042; Life Technologies, Carlsbad, CA) and Alexa Fluor 647 (A21245; Life Technologies). Slides were examined on a Zeiss AXIO Imager A1 & HBO100 (Zeiss, Oberkochen, Germany) using AxioVision software (Zeiss).

Statistical Analyses

All statistical analyses were performed using SPSS version 23.0 for Windows (IBM Corp., Armonk, NY). All P values of <0.05 were considered significant.

Participants’ baseline characteristics are summarized in Table 1. According to the glycemic status, age, BMI, and HOMA of insulin resistance (HOMA-IR) were increased, whereas HOMA of pancreatic β-cell function (HOMA-β) was decreased. CD8+CD57+ and CD8+CD28null T cells were significantly increased in patients with prediabetes and type 2 diabetes compared with those with NGT (Fig. 1A and B). However, senescent CD4+CD57+ or CD4+CD28null T-cell frequency was not significantly different (Fig. 1C and D). In Supplementary Fig. 2, representative flow cytometry plots present CD57 and CD28 expression in the CD8+ T-cell subset in the groups. In simple correlation analyses, fasting plasma glucose levels correlated significantly with frequency of CD8+CD57+ and CD8+CD28null T cells (Supplementary Table 1). There was a positive correlation between CD8+CD57+ T-cell frequency and HOMA-IR. Logistic regression analyses were conducted to determine odds ratios for predicting hyperglycemia (Table 2). After adjustment for traditional diabetes risk factors, frequencies of both CD8+CD57+ and CD8+CD28null T cells were significantly associated with hyperglycemia in the cross-sectional data set (N = 805). When divided into two groups based upon tertiles of CD8+CD57+ T-cell frequency in peripheral blood, HOMA-IR but not HOMA-β was significantly different in the higher (second to third) tertiles compared with the lower (first) tertile (Supplementary Table 2). When a cutoff of ≥2.34 for HOMA-IR was chosen (14), CD8+CD57+ T-cell frequencies were independently associated with insulin resistance (Supplementary Table 3).

Table 1

Baseline characteristics of the study participants according to glycemic status (N = 805)

Baseline characteristicsNGT (N = 222)Prediabetes (N = 302)Type 2 diabetes (N = 281)P value
Demographics     
 Age (years) 56.5 (48.0–67.0) 62.0 (54.0–69.0) 64.0 (58.0–70.0) <0.001 
 Male sex [n (%)] 125 (56.3) 215 (71.2) 184 (65.5) 0.002 
 BMI (kg/m224.5 (22.5–26.7) 25.5 (23.2–27.6) 25.6 (24.0–27.9) <0.001 
 Waist circumference (cm) 85.0 (79.0–91.3) 87.9 (82.0–93.3) 89.0 (83.0–94.0) <0.001 
 Hypertension [n (%)] 130 (58.6) 257 (85.1) 242 (86.1) <0.001 
 Systolic blood pressure (mmHg) 126.3 (114.8–136.5) 129.2 (120.0–139.0) 129.5 (119.0–141.3) 0.014 
 Diastolic blood pressure (mmHg) 79.0 ± 10.4 79.3 ± 10.6 77.3 ± 10.3 0.046 
 Hyperlipidemia [n (%)] 94 (42.3) 173 (57.3) 154 (54.8) 0.002 
 Coronary artery disease [n (%)] 51 (23.0) 141 (46.7) 151 (53.7) <0.001 
 Glucose-lowering drug use* [n (%)] — — 104 (37.0) — 
Laboratory indices     
 Fasting glucose (mg/dL) 92.0 (85.0–98.0) 99.0 (92.0–107.0) 117.0 (100.0–134.0) <0.001 
 HbA1c (%) 5.50 (5.40–5.70) 5.80 (5.60–6.00) 6.80 (6.30–7.30) <0.001 
 HbA1c (mmol/mol) 36.6 (35.5–38.8) 39.9 (37.7–42.1) 50.8 (45.4–56.3) <0.001 
 AST (IU/L) 21.0 (18.0–27.0) 23.0 (19.0–28.0) 23.0 (18.0–27.0) 0.030 
 ALT (IU/L) 16.0 (12.0–24.5) 20.0 (14.0–27.0) 19.0 (14.0–28.0) <0.001 
 BUN (mg/dL) 14.9 (12.6–17.7) 16.2 (13.4–19.9) 16.9 (13.6–20.2) <0.001 
 Creatinine (mg/dL) 0.89 (0.77–1.05) 0.92 (0.77–1.11) 0.97 (0.80–1.14) 0.053 
 eGFR MDRD (mL/min/1.73 m282.8 (70.8–96.3) 84.1 (66.9–98.9) 79.8 (65.1–92.9) 0.034 
 Total cholesterol (mg/dL) 175.0 (153.8–203.0) 164.0 (141.0–190.5) 155.5 (133.0–178.8) <0.001 
 Triglycerides (mg/dL) 101.5 (73.0–153.5) 115.0 (85.0–163.0) 119.0 (87.0–174.0) 0.002 
 HDL cholesterol (mg/dL) 51.0 (43.0–59.0) 46.0 (41.0–56.0) 46.0 (39.0–52.0) <0.001 
 LDL cholesterol (mg/dL) 102.0 (79.0–124.0) 87.1 (69.1–110.0) 81.0 (64.1–100.8) <0.001 
 Uric acid (mg/dL) 5.10 (4.10–6.15) 5.50 (4.50–6.60) 5.20 (4.30–6.30) 0.041 
 WBC count (103/µL) 5.95 (4.85–7.23) 6.19 (5.13–7.30) 6.42 (5.55–7.39) 0.012 
 HOMA-β (%) 141.0 ± 199.6 124.6 ± 91.6 85.6 ± 66.5 <0.001 
 HOMA-IR 2.19 ± 1.11 2.85 ± 1.97 3.38 ± 2.29 <0.001 
Frequency of T-cell subset     
 CD57+ cells in CD4+ T cells (%) 4.92 (3.10–7.37) 4.80 (3.40–7.23) 5.00 (3.10–7.83) 0.896 
 CD28null cells in CD4+ T cells (%) 3.00 (1.20–7.15) 3.50 (1.30–9.33) 3.66 (1.50–9.55) 0.227 
 CD57+ cells in CD8+ T cells (%) 38.7 ± 16.6 46.6 ± 16.1 47.8 ± 16.9 <0.001 
 CD28null cells in CD8+ T cells (%) 39.5 ± 19.5 48.2 ± 18.3 49.0 ± 18.2 <0.001 
Baseline characteristicsNGT (N = 222)Prediabetes (N = 302)Type 2 diabetes (N = 281)P value
Demographics     
 Age (years) 56.5 (48.0–67.0) 62.0 (54.0–69.0) 64.0 (58.0–70.0) <0.001 
 Male sex [n (%)] 125 (56.3) 215 (71.2) 184 (65.5) 0.002 
 BMI (kg/m224.5 (22.5–26.7) 25.5 (23.2–27.6) 25.6 (24.0–27.9) <0.001 
 Waist circumference (cm) 85.0 (79.0–91.3) 87.9 (82.0–93.3) 89.0 (83.0–94.0) <0.001 
 Hypertension [n (%)] 130 (58.6) 257 (85.1) 242 (86.1) <0.001 
 Systolic blood pressure (mmHg) 126.3 (114.8–136.5) 129.2 (120.0–139.0) 129.5 (119.0–141.3) 0.014 
 Diastolic blood pressure (mmHg) 79.0 ± 10.4 79.3 ± 10.6 77.3 ± 10.3 0.046 
 Hyperlipidemia [n (%)] 94 (42.3) 173 (57.3) 154 (54.8) 0.002 
 Coronary artery disease [n (%)] 51 (23.0) 141 (46.7) 151 (53.7) <0.001 
 Glucose-lowering drug use* [n (%)] — — 104 (37.0) — 
Laboratory indices     
 Fasting glucose (mg/dL) 92.0 (85.0–98.0) 99.0 (92.0–107.0) 117.0 (100.0–134.0) <0.001 
 HbA1c (%) 5.50 (5.40–5.70) 5.80 (5.60–6.00) 6.80 (6.30–7.30) <0.001 
 HbA1c (mmol/mol) 36.6 (35.5–38.8) 39.9 (37.7–42.1) 50.8 (45.4–56.3) <0.001 
 AST (IU/L) 21.0 (18.0–27.0) 23.0 (19.0–28.0) 23.0 (18.0–27.0) 0.030 
 ALT (IU/L) 16.0 (12.0–24.5) 20.0 (14.0–27.0) 19.0 (14.0–28.0) <0.001 
 BUN (mg/dL) 14.9 (12.6–17.7) 16.2 (13.4–19.9) 16.9 (13.6–20.2) <0.001 
 Creatinine (mg/dL) 0.89 (0.77–1.05) 0.92 (0.77–1.11) 0.97 (0.80–1.14) 0.053 
 eGFR MDRD (mL/min/1.73 m282.8 (70.8–96.3) 84.1 (66.9–98.9) 79.8 (65.1–92.9) 0.034 
 Total cholesterol (mg/dL) 175.0 (153.8–203.0) 164.0 (141.0–190.5) 155.5 (133.0–178.8) <0.001 
 Triglycerides (mg/dL) 101.5 (73.0–153.5) 115.0 (85.0–163.0) 119.0 (87.0–174.0) 0.002 
 HDL cholesterol (mg/dL) 51.0 (43.0–59.0) 46.0 (41.0–56.0) 46.0 (39.0–52.0) <0.001 
 LDL cholesterol (mg/dL) 102.0 (79.0–124.0) 87.1 (69.1–110.0) 81.0 (64.1–100.8) <0.001 
 Uric acid (mg/dL) 5.10 (4.10–6.15) 5.50 (4.50–6.60) 5.20 (4.30–6.30) 0.041 
 WBC count (103/µL) 5.95 (4.85–7.23) 6.19 (5.13–7.30) 6.42 (5.55–7.39) 0.012 
 HOMA-β (%) 141.0 ± 199.6 124.6 ± 91.6 85.6 ± 66.5 <0.001 
 HOMA-IR 2.19 ± 1.11 2.85 ± 1.97 3.38 ± 2.29 <0.001 
Frequency of T-cell subset     
 CD57+ cells in CD4+ T cells (%) 4.92 (3.10–7.37) 4.80 (3.40–7.23) 5.00 (3.10–7.83) 0.896 
 CD28null cells in CD4+ T cells (%) 3.00 (1.20–7.15) 3.50 (1.30–9.33) 3.66 (1.50–9.55) 0.227 
 CD57+ cells in CD8+ T cells (%) 38.7 ± 16.6 46.6 ± 16.1 47.8 ± 16.9 <0.001 
 CD28null cells in CD8+ T cells (%) 39.5 ± 19.5 48.2 ± 18.3 49.0 ± 18.2 <0.001 

Continuous variables are described as mean ± SD for parametric variables and median (interquartile range) for nonparametric variables unless otherwise indicated. The P values were calculated using the Kruskal-Wallis test or one-way ANOVA for continuous variables and χ2 tests for categorical variables. BUN, blood urea nitrogen; eGFR, estimated glomerular filtration rate; WBC, white blood cell.

*Oral hypoglycemic agents or insulin. HOMA-β = (fasting serum insulin [µU/mL] × 20)/(fasting serum glucose [mmol/L] − 3.5); HOMA-IR = (fasting serum insulin [µU/mL] × fasting serum glucose [mmol/L])/22.5.

Figure 1

The relative frequency of senescent T cells in the peripheral blood from subjects with NGT (N = 222), prediabetes (N = 302), and type 2 diabetes (N = 281). Both CD57+ and CD28null fractions are significantly increased in CD8+ T cells of participants with prediabetes and type 2 diabetes (A and B) but not in CD4+ T cells (C and D). The horizontal line in the middle of each box indicates the median, the top and bottom borders of the box mark the 75th and 25th percentiles, respectively, and the top and bottom whiskers mark the maximum and minimum values, respectively.

Figure 1

The relative frequency of senescent T cells in the peripheral blood from subjects with NGT (N = 222), prediabetes (N = 302), and type 2 diabetes (N = 281). Both CD57+ and CD28null fractions are significantly increased in CD8+ T cells of participants with prediabetes and type 2 diabetes (A and B) but not in CD4+ T cells (C and D). The horizontal line in the middle of each box indicates the median, the top and bottom borders of the box mark the 75th and 25th percentiles, respectively, and the top and bottom whiskers mark the maximum and minimum values, respectively.

Close modal
Table 2

Multiple logistic regression models for independent determinants of the presence of hyperglycemia (N = 805)

VariablesUnivariate
Multivariate
OR95% CIP valueOR95% CIP value
Model 1 for CD8+CD57+ T cells       
 CD8+CD57+ T cells (%) 1.697 1.436–2.006 <0.001 1.415 1.160–1.727 0.001 
 Age, per year 1.052 1.036–1.067 <0.001 1.018 1.000–1.037 0.052 
 Sex (0 = male, 1 = female) 0.594 0.433–0.816 0.001 0.723 0.493–1.059 0.096 
 BMI (kg/m21.114 1.059–1.172 <0.001 1.074 1.015–1.136 0.013 
 Hypertension (0 = no, 1 = yes) 4.204 2.953–5.985 <0.001 3.686 2.409–5.641 <0.001 
 Hyperlipidemia (0 = no, 1 = yes) 1.739 1.273–2.377 0.001 1.399 0.974–2.010 0.069 
 Coronary artery disease (0 = no, 1 = yes) 3.364 2.365–4.786 <0.001 3.838 2.542–5.796 <0.001 
 WBC count (103/µL) 1.135 1.030–1.251 0.010 1.090 0.980–1.213 0.114 
Model 2 for CD8+CD28null T cells       
 CD8+CD28null T cells (%) 1.671 1.411–1.979 <0.001 1.384 1.145–1.674 0.001 
 Age, per year 1.052 1.036–1.067 <0.001 1.022 1.004–1.040 0.017 
 Sex (0 = male, 1 = female) 0.594 0.433–0.816 0.001 0.648 0.446–0.942 0.023 
 BMI (kg/m21.114 1.059–1.172 <0.001 1.073 1.014–1.135 0.014 
 Hypertension (0 = no, 1 = yes) 4.204 2.953–5.985 <0.001 3.557 2.319–5.456 <0.001 
 Hyperlipidemia (0 = no, 1 = yes) 1.739 1.273–2.377 0.001 1.349 0.940–1.936 0.105 
 Coronary artery disease (0 = no, 1 = yes) 3.364 2.365–4.786 <0.001 3.827 2.539–5.768 <0.001 
 WBC count (103/µL) 1.135 1.030–1.251 0.010 1.081 0.972–1.203 0.152 
VariablesUnivariate
Multivariate
OR95% CIP valueOR95% CIP value
Model 1 for CD8+CD57+ T cells       
 CD8+CD57+ T cells (%) 1.697 1.436–2.006 <0.001 1.415 1.160–1.727 0.001 
 Age, per year 1.052 1.036–1.067 <0.001 1.018 1.000–1.037 0.052 
 Sex (0 = male, 1 = female) 0.594 0.433–0.816 0.001 0.723 0.493–1.059 0.096 
 BMI (kg/m21.114 1.059–1.172 <0.001 1.074 1.015–1.136 0.013 
 Hypertension (0 = no, 1 = yes) 4.204 2.953–5.985 <0.001 3.686 2.409–5.641 <0.001 
 Hyperlipidemia (0 = no, 1 = yes) 1.739 1.273–2.377 0.001 1.399 0.974–2.010 0.069 
 Coronary artery disease (0 = no, 1 = yes) 3.364 2.365–4.786 <0.001 3.838 2.542–5.796 <0.001 
 WBC count (103/µL) 1.135 1.030–1.251 0.010 1.090 0.980–1.213 0.114 
Model 2 for CD8+CD28null T cells       
 CD8+CD28null T cells (%) 1.671 1.411–1.979 <0.001 1.384 1.145–1.674 0.001 
 Age, per year 1.052 1.036–1.067 <0.001 1.022 1.004–1.040 0.017 
 Sex (0 = male, 1 = female) 0.594 0.433–0.816 0.001 0.648 0.446–0.942 0.023 
 BMI (kg/m21.114 1.059–1.172 <0.001 1.073 1.014–1.135 0.014 
 Hypertension (0 = no, 1 = yes) 4.204 2.953–5.985 <0.001 3.557 2.319–5.456 <0.001 
 Hyperlipidemia (0 = no, 1 = yes) 1.739 1.273–2.377 0.001 1.349 0.940–1.936 0.105 
 Coronary artery disease (0 = no, 1 = yes) 3.364 2.365–4.786 <0.001 3.827 2.539–5.768 <0.001 
 WBC count (103/µL) 1.135 1.030–1.251 0.010 1.081 0.972–1.203 0.152 

OR, odds ratio; WBC, white blood cell.

Of 149 NGT participants, 58 developed prediabetes or type 2 diabetes during follow-up (2.32 ± 1.45 years). NGT participants were divided into two groups based upon tertiles of CD8+CD57+ or CD8+CD28null T-cell frequency: low (first to second tertile) and high (third tertile). In the high CD8+CD57+ T-cell group, 57% (20 out of 35) developed hyperglycemia compared with 33% (38 out of 114) in the low group. We observed a significant difference in incidence rates among participants with a high versus low frequency of CD8+CD57+ T cells (Kaplan-Meier and log-rank test, P = 0.001) (Fig. 2A). However, this difference was not found for CD8+CD28null T cells (Fig. 2B). A Cox regression model (Fig. 2C) revealed that, after adjusting for traditional risk factors for diabetes, the hazard ratio for developing hyperglycemia per percentage of CD8+CD57+ T cells was 1.785 (95% CI 1.298–2.455).

Figure 2

Increased frequency of CD8+CD57+ T cells predicts development of hyperglycemia in participants with NGT (N = 149). Kaplan-Meier curves for hyperglycemia development according to the highest tertile vs. the lower two tertiles of the frequency of CD8+CD57+ (A) or CD8+CD28null (B) T cells. C: Cox regression analyses for incidence of hyperglycemia. Model 1, unadjusted; model 2, adjusted for age and sex; model 3, adjusted for age, sex, BMI, hypertension, hyperlipidemia, coronary artery disease, and white blood cell count.

Figure 2

Increased frequency of CD8+CD57+ T cells predicts development of hyperglycemia in participants with NGT (N = 149). Kaplan-Meier curves for hyperglycemia development according to the highest tertile vs. the lower two tertiles of the frequency of CD8+CD57+ (A) or CD8+CD28null (B) T cells. C: Cox regression analyses for incidence of hyperglycemia. Model 1, unadjusted; model 2, adjusted for age and sex; model 3, adjusted for age, sex, BMI, hypertension, hyperlipidemia, coronary artery disease, and white blood cell count.

Close modal

In humans, CMV is known as an important antigen for repetitive T-cell stimulation and is involved in the accumulation of CD28null or CD57+ senescent T cells (9). Therefore, we analyzed CMV-specific antigen reactivity of T cells. Proinflammatory and cytotoxic functions of T cells were evaluated and compared among 56 baseline NGT participants who did versus did not develop hyperglycemia (Supplementary Table 4). All participants were seropositive for CMV in the current study (data not shown). CMV pp65-specific CD107a-expressing CD8+ T cells, which represent degranulation of cytotoxic proteins such as perforin and granzymes (15), were more frequently observed in those who did compared with those who did not progress to hyperglycemia.

Adipose tissue is a major target of insulin action and immune cell reservoir (16). Using immunohistochemistry, we identified infiltrated CD8+ T cells adjacent to macrophages within adipose tissues (Supplementary Fig. 3A). Furthermore, immunofluorescent analyses showed histological evidence of greater CD8+CD57+ T-cell infiltration in omental adipose tissues of patients with prediabetes or type 2 diabetes compared with those with NGT (Supplementary Fig. 3B and C).

The current study demonstrates that increased senescent CD8+ T cells in peripheral blood are independently associated with prevalence and incidence of prediabetes or type 2 diabetes. Furthermore, increased frequency of CD8+CD57+ T cells was associated with insulin resistance, and we observed histological evidence of CD8+CD57+ T-cell infiltration in visceral adipose tissues (VATs) of patients with prediabetes or type 2 diabetes.

Although type 2 diabetes pathogenesis is not fully understood, insulin resistance is the hallmark (17). Obesity is associated with chronic low-grade inflammation in VATs and a sustained whole-body proinflammatory state (18). Infiltration of T cells into adipose tissue has been extensively reported (19,20). Nishimura et al. (21) found large numbers of CD8+ effector T cells in epididymal adipose tissue in mice fed a high-fat diet. Immunologic/genetic depletion of CD8+ T cells lowered macrophage infiltration and adipose tissue inflammation and ameliorated systemic insulin resistance. In a human study that included patients with diabetes, CD4+ lymphocytes in VAT biopsies correlated significantly with BMI (19). In the current study, senescent CD8+ T cells and not CD4+ T cells were significantly associated with present and future hyperglycemia.

Common persistent viral infections (especially human CMV) are attributed to increasing CD8+CD28null (CD8+CD57+) T-cell populations with age (9) and have been shown to express perforin, granzymes, and granulysin, with high cytotoxic potential (8,22,23). This finding aligns with our previous observation that CMV pp65-specific interferon-γ–, tumor necrosis factor-α–, and CD107a-expressing cells were more frequently observed in CD8+CD57+ T cells compared with CD8+CD57 T cells (24). Previous studies demonstrated that proinflammatory cytokines could promote insulin resistance (25). Correspondingly, we found that increased frequency of CD8+CD57+ T cells was independently associated with insulin resistance measured by HOMA-IR. In addition, among participants with baseline NGT, CMV pp65-specific CD107a-expressing CD8+ T cells were more frequently observed in those who did versus did not progress to hyperglycemia. How senescent T cells interact with other immune cells within target tissues (including adipose) in hyperglycemia development remains to be investigated. Although our findings suggest that senescent T cells might be involved in the early development of diabetes, we were unable to elucidate whether senescent T cells are indicators or pathogenic players for hyperglycemia.

In conclusion, present and future hyperglycemia (prediabetes or type 2 diabetes) were associated with increased senescent CD8+ T cells in peripheral blood. Larger prospective studies and sophisticated mechanistic experiments are warranted to confirm whether modulation of immunosenescence is a potential new therapeutic target of diabetes.

Funding. This work was supported by grants from the Korean Health Technology R&D Project, Ministry of Health and Welfare, Republic of Korea (HI13C0715 to S.P. and HI17C0913 to Y.-h.L.) and by the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Science, ICT and Future Planning (NRF-2015R1A2A2A01007346 to S.P. and NRF-2016R1A5A1010764 to Y.-h.L.). This work was also supported by the KAIST Future Systems Healthcare Project from the Ministry of Science, ICT and Future Planning.

Duality of Interest. No potential conflicts of interest relevant to this article were reported.

Author Contributions. Y.-h.L. and S.R.K. wrote the manuscript, analyzed data, and performed the statistical analysis. D.H.H., Y.D.H., J.H.K., and S.H.K. contributed to acquisition of data. H.T.Y., C.J.L., and K.H.K. analyzed data and performed the statistical analysis. B.-H.M. and D.-H.K. performed histologic analyses. J.W.C. provided critical review, advice, and consultation throughout. Y.-h.L., S.R.K., D.H.H., H.T.Y., Y.D.H., J.H.K., S.H.K., C.J.L., B.-H.M., D.-H.K., K.H.K., J.W.C., W.-W.L., E.-C.S., and S.P. contributed to critical revision of the manuscript and read and approved the final submitted version of the manuscript. W.-W.L., E.-C.S., and S.P. are the guarantors of this work and, as such, had full access to all of the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis.

1.
Franceschi
C
,
Bonafè
M
,
Valensin
S
, et al
.
Inflamm-aging. An evolutionary perspective on immunosenescence
.
Ann N Y Acad Sci
2000
;
908
:
244
254
[PubMed]
2.
Maeda
T
,
Yamada
H
,
Nagamine
R
, et al
.
Involvement of CD4+,CD57+ T cells in the disease activity of rheumatoid arthritis
.
Arthritis Rheum
2002
;
46
:
379
384
[PubMed]
3.
Grahame-Clarke
C
,
Chan
NN
,
Andrew
D
, et al
.
Human cytomegalovirus seropositivity is associated with impaired vascular function
.
Circulation
2003
;
108
:
678
683
[PubMed]
4.
Kaplan
RC
,
Sinclair
E
,
Landay
AL
, et al
.
T cell activation and senescence predict subclinical carotid artery disease in HIV-infected women
.
J Infect Dis
2011
;
203
:
452
463
[PubMed]
5.
Bergström
I
,
Backteman
K
,
Lundberg
A
,
Ernerudh
J
,
Jonasson
L
.
Persistent accumulation of interferon-γ-producing CD8+CD56+ T cells in blood from patients with coronary artery disease
.
Atherosclerosis
2012
;
224
:
515
520
[PubMed]
6.
Olson
NC
,
Doyle
MF
,
Jenny
NS
, et al
.
Decreased naive and increased memory CD4(+) T cells are associated with subclinical atherosclerosis: the multi-ethnic study of atherosclerosis
.
PLoS One
2013
;
8
:
e71498
[PubMed]
7.
Giubilato
S
,
Liuzzo
G
,
Brugaletta
S
, et al
.
Expansion of CD4+CD28null T-lymphocytes in diabetic patients: exploring new pathogenetic mechanisms of increased cardiovascular risk in diabetes mellitus
.
Eur Heart J
2011
;
32
:
1214
1226
[PubMed]
8.
Youn
JC
,
Yu
HT
,
Lim
BJ
, et al
.
Immunosenescent CD8+ T cells and C-X-C chemokine receptor type 3 chemokines are increased in human hypertension
.
Hypertension
2013
;
62
:
126
133
[PubMed]
9.
Strioga
M
,
Pasukoniene
V
,
Characiejus
D
.
CD8+ CD28- and CD8+ CD57+ T cells and their role in health and disease
.
Immunology
2011
;
134
:
17
32
[PubMed]
10.
Dumitriu
IE
,
Araguás
ET
,
Baboonian
C
,
Kaski
JC
.
CD4+ CD28 null T cells in coronary artery disease: when helpers become killers
.
Cardiovasc Res
2009
;
81
:
11
19
[PubMed]
11.
Stern
MP
.
Diabetes and cardiovascular disease. The “common soil” hypothesis
.
Diabetes
1995
;
44
:
369
374
[PubMed]
12.
American Diabetes Association
.
2. Classification and diagnosis of diabetes
.
Diabetes Care
2017
;
40
(
Suppl. 1
):
S11
S24
[PubMed]
13.
Olson
DE
,
Rhee
MK
,
Herrick
K
,
Ziemer
DC
,
Twombly
JG
,
Phillips
LS
.
Screening for diabetes and pre-diabetes with proposed A1C-based diagnostic criteria
.
Diabetes Care
2010
;
33
:
2184
2189
[PubMed]
14.
Lee
S
,
Choi
S
,
Kim
HJ
, et al
.
Cutoff values of surrogate measures of insulin resistance for metabolic syndrome in Korean non-diabetic adults
.
J Korean Med Sci
2006
;
21
:
695
700
[PubMed]
15.
Alter
G
,
Malenfant
JM
,
Altfeld
M
.
CD107a as a functional marker for the identification of natural killer cell activity
.
J Immunol Methods
2004
;
294
:
15
22
[PubMed]
16.
Wellen
KE
,
Hotamisligil
GS
.
Obesity-induced inflammatory changes in adipose tissue
.
J Clin Invest
2003
;
112
:
1785
1788
[PubMed]
17.
Defronzo
RA
.
Banting lecture. From the triumvirate to the ominous octet: a new paradigm for the treatment of type 2 diabetes mellitus
.
Diabetes
2009
;
58
:
773
795
[PubMed]
18.
Després
JP
.
Body fat distribution and risk of cardiovascular disease: an update
.
Circulation
2012
;
126
:
1301
1313
[PubMed]
19.
Kintscher
U
,
Hartge
M
,
Hess
K
, et al
.
T-lymphocyte infiltration in visceral adipose tissue: a primary event in adipose tissue inflammation and the development of obesity-mediated insulin resistance
.
Arterioscler Thromb Vasc Biol
2008
;
28
:
1304
1310
[PubMed]
20.
Duffaut
C
,
Zakaroff-Girard
A
,
Bourlier
V
, et al
.
Interplay between human adipocytes and T lymphocytes in obesity: CCL20 as an adipochemokine and T lymphocytes as lipogenic modulators
.
Arterioscler Thromb Vasc Biol
2009
;
29
:
1608
1614
[PubMed]
21.
Nishimura
S
,
Manabe
I
,
Nagasaki
M
, et al
.
CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity
.
Nat Med
2009
;
15
:
914
920
[PubMed]
22.
Fiorentino
S
,
Dalod
M
,
Olive
D
,
Guillet
JG
,
Gomard
E
.
Predominant involvement of CD8+CD28- lymphocytes in human immunodeficiency virus-specific cytotoxic activity
.
J Virol
1996
;
70
:
2022
2026
[PubMed]
23.
Le Priol
Y
,
Puthier
D
,
Lécureuil
C
, et al
.
High cytotoxic and specific migratory potencies of senescent CD8+ CD57+ cells in HIV-infected and uninfected individuals
.
J Immunol
2006
;
177
:
5145
5154
[PubMed]
24.
Yu
HT
,
Youn
JC
,
Kim
JH
, et al
.
Arterial stiffness is associated with cytomegalovirus-specific senescent CD8+ T cells
.
J Am Heart Assoc
2017
;
6
:
1
14
[PubMed]
25.
Zinman
B
,
Hanley
AJ
,
Harris
SB
,
Kwan
J
,
Fantus
IG
.
Circulating tumor necrosis factor-alpha concentrations in a native Canadian population with high rates of type 2 diabetes mellitus
.
J Clin Endocrinol Metab
1999
;
84
:
272
278
[PubMed]
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at http://www.diabetesjournals.org/content/license.

Supplementary data