Abstract
Although CD4+CD25+FOXP3+ regulatory T (TREG) cells have been studied in patients with COVID-19, changes in the TREG cell population have not been longitudinally examined during the course of COVID-19. In this study, we longitudinally investigated the quantitative and qualitative changes in the TREG cell population in patients with COVID-19. We found that the frequencies of total TREG cells and CD45RA−FOXP3hi activated TREG cells were significantly increased 15–28 d postsymptom onset in severe patients, but not in mild patients. TREG cells from severe patients exhibited not only increased proliferation but also enhanced apoptosis, suggesting functional derangement of the TREG cell population during severe COVID-19. The suppressive functions of the TREG cell population did not differ between patients with severe versus mild COVID-19. The frequency of TREG cells inversely correlated with SARS-CoV-2–specific cytokine production by CD4+ T cells and their polyfunctionality in patients with mild disease, suggesting that TREG cells are major regulators of virus-specific CD4+ T cell responses during mild COVID-19. However, such correlations were not observed in patients with severe disease. Thus, in this study, we describe distinctive changes in the TREG cell population in patients with severe and mild COVID-19. Our study provides a deep understanding of host immune responses upon SARS-CoV-2 infection in regard to TREG cells.
Introduction
Since the first report of COVID-19 caused by SARS-CoV-2 infection in December 2019 in Wuhan, China (1, 2), 6 million cases of mortality had been reported globally as of April 10, 2022 (3). SARS-CoV-2 infection manifests as various forms of clinical illness, from asymptomatic infection to severe/critical illness (4, 5). Severe/critical COVID-19 is mediated by exaggerated inflammatory responses and accompanied by acute respiratory distress syndrome and multiple organ dysfunction.
Since the emergence of COVID-19, many studies have examined the host immune responses against SARS-CoV-2, including neutralizing Abs, CD4+ and CD8+ T cell responses (6–12), and inflammatory responses, including hyperactivation of monocytes/macrophages and neutrophils and the production of proinflammatory cytokines (13–16). However, the changes in suppressive immune cells during COVID-19 remain to be fully elucidated.
CD4+CD25+FOXP3+ regulatory T (TREG) cells are representative suppressive immune cells that maintain homeostasis in the immune system (17–19). TREG cells exert immunosuppressive functions via CTLA-4, which competes with the CD28 expressed by effector T cells for binding to B7 (20–22), and immunosuppressive cytokines, such as IL-10, IL-35, and TGF-β (23–26). In humans, the CD4+CD25+FOXP3+ TREG cell population includes distinct subpopulations, such as CD45RA−FOXP3hi activated TREG cells and CD45RA+FOXP3lo resting TREG cells (27). CD45RA−FOXP3hi activated TREG cells exert stronger suppressive functions than CD45RA+FOXP3lo resting TREG cells but are vulnerable to apoptosis (27, 28).
Changes in the TREG cell population have been reported in various viral diseases, particularly viral hepatitis. In patients with hepatitis C virus (HCV) infection, the frequency of TREG cells increases in the peripheral blood (PB) and liver (29–31), and the increase in TREG cell frequency has been related to CD8+ T cell dysfunction and high viral load, indicating that TREG cells contribute to viral persistence by suppressing T cell responses during HCV infection. In contrast, the frequency of TREG cells decreases in the PB via Fas-mediated apoptosis in patients with hepatitis A virus (HAV) infection. A decreased TREG cell frequency is associated with severe liver injury (32), which is mediated by an immune-mediated mechanism during HAV infection (33), indicating a protective role of TREG cells against immune-mediated host injury during viral infection.
Dual roles of TREG cells in viral infection have also been demonstrated in mouse models. Depletion of TREG cells results in enhanced CD8+ T cell responses against infecting viruses and accelerated viral clearance in mouse models of infection with HSV (34) or lymphocytic choriomeningitis virus (35), indicating that TREG cells control virus-specific cellular immune responses during viral infection. In contrast, depletion of TREG cells results in greater disease severity because of an increased immunopathological reaction in mouse models of corneal infection with HSV (36) or infection with respiratory syncytial virus or West Nile virus (37–39), supporting host protective roles of TREG cells during viral infection. These previous findings show both immunosuppressive and host protective roles of TREG cells during viral infection (40–42).
Perturbations in the TREG cell population were recently reported among patients with COVID-19 (43–45). However, this cross-sectional study has not directly investigated suppressive functions of TREG cells and relationships between TREG cells and effector T cell functions. In this study, we examined the quantitative and qualitative changes in the TREG cell population in patients with COVID-19 and healthy donors. We analyzed the frequency of the TREG cell population and subpopulation and their phenotypes during the course of COVID-19 using longitudinal PB samples. We also compared the frequency, phenotype, and suppressive function of TREG cells among patients with severe and mild COVID-19 and healthy donors and found that the TREG cell population is distinctively modulated in patients with severe and mild disease.
Materials and Methods
Study design
For this study, we enrolled PCR-confirmed COVID-19 patients in the year 2020 with severe (n = 21; male/female ratio = 13:8; mean age = 62.9 y, range = 41–81 y) or mild (n = 35; male/female ratio = 16:19; mean age = 47.8 y, range = 18–96 y) disease. SARS-CoV-2 infection was confirmed using the PowerChek2019-nCoV Real-time PCR Kit (Kogene Biotech) at Chungnam National University Hospital (28 patients) or Allplex SARS-CoV-2 Assay (Seegene) at Chungbuk National University Hospital (28 patients; Supplemental Table I). The day of symptom onset could be defined in each patient, and thus days postsymptom onset (DPSO) could be determined for each sampling date. Three to four follow-up PB samples were obtained from each patient. The mild patients included patients with mild and moderate symptoms at peak severity, and severe patients included severe and critical patients according to the National Institutes of Health severity scale (46). PB samples from 23 SARS-CoV-2–unexposed healthy donors were used as controls. Clinical information of each patient, including the date of sampling, comorbidities, and treatment during COVID-19, is presented in Supplemental Table I. We also enrolled healthy donors (n = 23; mean age = 39.9 y, range = 24–62 y). This study was reviewed and approved by the institutional review board of all participating institutions and conducted according to the principles of the Declaration of Helsinki. Informed consent was obtained from all donors and patients.
PBMCs were isolated from whole blood by density gradient centrifugation using Lymphocyte Separation Medium (Corning).
Flow cytometry
Total PBMCs were incubated at room temperature for 15 min with fluorochrome-conjugated Abs against cell surface markers. To exclude dead cells, we used a Live/Dead Cell Stain Kit (Invitrogen). For intracellular staining, fixation and permeabilization were achieved with the Foxp3/Transcription Factor Staining Buffer Kit (eBioscience) for 15 min, and Abs specific for Foxp3, Ki-67, and CTLA-4 were added for another 15 min (Table I). An LSR II instrument (BD Bioscience) was used for flow cytometry. The gating strategy for phenotyping the TREG cell population is presented in Supplemental Fig. 1A.
Name of Abs . | Company . | Cat# . |
---|---|---|
BV421, anti-human CD25, clone M-A251 | BD | 562442 |
BV510, anti-human CD4, clone SK3 | BD | 562970 |
BV605, anti-human CD3, clone UCHT1 | BD | 742623 |
BV786, anti-human Ki-67, clone B56 | BD | 563756 |
PE, anti-human Foxp3, clone PCH101 | eBioscience | 12-4776-42 |
PE-TR, anti-human CD14, clone 61D3 | eBioscience | 61-0149-42 |
PE-TR, anti-human CD19, clone HIB19 | eBioscience | 61-0199-42 |
PerCP-Cy5.5, anti-human CD127, clone HIL-7R-M21 | BD | 560551 |
Allophycocyanin, anti-human CTLA-4, clone BNI3 | BD | 555855 |
Allophycocyanin-H7, anti-human CD45RA, clone HI100 | BioLegend | 304128 |
BV711, anti-human CD25, clone 2A3 | BD | 563159 |
BV786, anti-human CD127, clone HIL-7R-M21 | BD | 563324 |
FITC, anti-human CD4, clone RPA-T4 | BD | 561842 |
PE-Cy7, anti-human Foxp3, clone PCH101 | eBioscience | 25-4776-42 |
PE, Annexin V | BD | 556422 |
Allophycocyanin, anti-human active caspase-3, clone C92-605 | BD | 560626 |
BV711, anti-human Ki-67, clone Ki-67 | BioLegend | 350516 |
BV786, anti-human CD4, clone SK3 | BD | 563877 |
FITC, anti-human CD8, clone RPA-T8 | BD | 555366 |
Allophycocyanin, anti-human CD3, clone UCHT1 | BioLegend | 300439 |
BV510, anti-human CD3, clone UCHT1 | BD | 563109 |
BV711, anti-human IFNg, clone B27 | BD | 564039 |
BV786, anti-human CD4, clone SK3 | BD | 563877 |
PerCP-Cy5.5, anti-human CD8, clone RPA-T8 | BD | 560662 |
PE-Cy7, anti-human TNF, clone Mab11 | eBioscience | 25-7349-82 |
Allophycocyanin, anti-human IL-2, clone MQ1-17H12 | BD | 554567 |
Allophycocyanin-H7, anti-human CD127, clone A019D5 | BioLegend | 351348 |
Name of Abs . | Company . | Cat# . |
---|---|---|
BV421, anti-human CD25, clone M-A251 | BD | 562442 |
BV510, anti-human CD4, clone SK3 | BD | 562970 |
BV605, anti-human CD3, clone UCHT1 | BD | 742623 |
BV786, anti-human Ki-67, clone B56 | BD | 563756 |
PE, anti-human Foxp3, clone PCH101 | eBioscience | 12-4776-42 |
PE-TR, anti-human CD14, clone 61D3 | eBioscience | 61-0149-42 |
PE-TR, anti-human CD19, clone HIB19 | eBioscience | 61-0199-42 |
PerCP-Cy5.5, anti-human CD127, clone HIL-7R-M21 | BD | 560551 |
Allophycocyanin, anti-human CTLA-4, clone BNI3 | BD | 555855 |
Allophycocyanin-H7, anti-human CD45RA, clone HI100 | BioLegend | 304128 |
BV711, anti-human CD25, clone 2A3 | BD | 563159 |
BV786, anti-human CD127, clone HIL-7R-M21 | BD | 563324 |
FITC, anti-human CD4, clone RPA-T4 | BD | 561842 |
PE-Cy7, anti-human Foxp3, clone PCH101 | eBioscience | 25-4776-42 |
PE, Annexin V | BD | 556422 |
Allophycocyanin, anti-human active caspase-3, clone C92-605 | BD | 560626 |
BV711, anti-human Ki-67, clone Ki-67 | BioLegend | 350516 |
BV786, anti-human CD4, clone SK3 | BD | 563877 |
FITC, anti-human CD8, clone RPA-T8 | BD | 555366 |
Allophycocyanin, anti-human CD3, clone UCHT1 | BioLegend | 300439 |
BV510, anti-human CD3, clone UCHT1 | BD | 563109 |
BV711, anti-human IFNg, clone B27 | BD | 564039 |
BV786, anti-human CD4, clone SK3 | BD | 563877 |
PerCP-Cy5.5, anti-human CD8, clone RPA-T8 | BD | 560662 |
PE-Cy7, anti-human TNF, clone Mab11 | eBioscience | 25-7349-82 |
Allophycocyanin, anti-human IL-2, clone MQ1-17H12 | BD | 554567 |
Allophycocyanin-H7, anti-human CD127, clone A019D5 | BioLegend | 351348 |
Cat#, catalog number.
Total blood lymphocyte counts were obtained by clinical laboratories at the time of blood sampling. We calculated the absolute counts of total CD3+ and CD4+ cells, TREG cells, activated TREG cells, and CTLA-4+ TREG cells based on the blood lymphocyte counts and the percentages of each cell type obtained from flow cytometry.
Reanalysis of single-cell RNA sequencing data
Recently published publicly available datasets were used in the reanalysis (47) (GSE152522; https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE152522). The original datasets were obtained by Ag-reactive T cell enrichment. In this assay, SARS-CoV-2–reactive CD4+ T cells were enriched based on the expression of surface activation-induced markers (CD137 and CD69) after ex vivo stimulation of PBMCs for 24 h with overlapping peptide (OLP) pools targeting the immunogenic domains of the spike and membrane proteins of SARS-CoV-2 from 30 acute COVID-19 patients (mild, n = 21; severe, n = 9). After downloading, the datasets were preprocessed and analyzed using the Seurat package (48). To correct the batch effect according to donor origin, dimensional reduction was performed using the mutual nearest neighbor (MNN) algorithm with 2000 variable genes (49) (RunFastMNN function). Uniform manifold approximation and projection (UMAP) was performed with the top MNN components (total CD4+ T cells, dimensions = 20; TREG cells, dimensions = 10) for cell clustering and visualization (RunUMAP and FindNeighbors function). Next, the cells were clustered by an unsupervised clustering method (total CD4+ T cells, resolution = 0.1; TREG cell resolution = 0.2; FindClusters function).
Differentially expressed genes (DEGs) were calculated based on the Wilcoxon rank sum test with default parameters (FindAllMarkers function). To characterize the immunophenotype of each cluster, we carried out gene set enrichment analysis (GSEA) with publicly available gene sets, including Gene Ontology: Biological Process database, using the piano package (50). Ontology terms related to immune responses were filtered using the following inclusion criteria: “T_CELL,” “IMMUNE,” “INNATE,” “ADAPTIVE,” “INFLAM,” “INTERLEUKIN,” “INTERFERON,” “NECROSIS,” “APOPTOSIS,” “SENESCENCE,” “NATURAL,” “LYMPHOCYTE,” “LEUKOCYTE,” “TRANSFORMING,” “CHEMO,” “CYTOTO,” “CYTOKINE,” “ANTIGEN,” and “SIGNALING.” To evaluate apoptosis-associated gene set enrichment, cells with active gene sets were identified using the AUCell package (51). The subcluster proportion was calculated in patients (mild, n = 15; severe, n = 7) whose TREG cell numbers were >50.
Anti-CD3/CD28 stimulation of PBMCs
PBMCs were stimulated with soluble anti-CD3 (0.1 μg/ml) and anti-CD28 (1 μg/ml) mAbs and cultured for 6 h at 37°C. After stimulation, apoptotic cells were stained with fluorochrome-conjugated annexin V Ab and analyzed by flow cytometry.
Apoptosis assay
To evaluate apoptotic cells among TREG cells, we stained PBMCs using a Live/Dead Cell Stain Kit and fluorochrome-conjugated Abs against surface markers, CD3, CD4, CD25, and CD127. After washing with 1× binding buffer, cells were stained with PE-conjugated annexin V (BD Biosciences). Fixation and permeabilization were performed before Foxp3 and active caspase-3 staining (Table I). The percentages of late apoptotic (annexin V+Live/Dead dye+) and active caspase-3+ cells were analyzed by flow cytometry in the gate of TREG cells (CD4+CD25+CD127loFOXP3+) or non-TREG cells (CD4+CD25−) T cells.
TREG cell depletion and suppression assay
Cryopreserved PBMCs were incubated for 12 h at 37°C after thawing. To deplete CD25+ TREG cells from PBMCs, we attached CD25 Microbeads II (Miltenyi Biotec) to total PBMCs, and labeled cells were depleted by an LD column (Miltenyi Biotec) and MACS separator (Miltenyi Biotec). Unlabeled cells that flew through the LD column were collected and used as CD25+ cell–depleted PBMCs. The magnetically labeled CD25+ cells remaining in the LD column were flushed out using a plunger. CD25+ cell–depleted PBMCs were labeled with 5 μM CellTrace Violet (CTV; CellTrace Violet Cell Proliferation Kit; Invitrogen). CD25+ cell–depleted, CTV-labeled PBMCs, serving as responder T (TRESP), were stimulated with soluble anti-CD3 (0.1 μg/ml; BD Pharmingen) and anti-CD28 (0.1 μg/ml; BD Pharmingen) with or without readdition of the depleted CD25+ cells. After 96 h of culture, proliferation of CD4+ and CD8+ TRESP cells was analyzed by assessing the percentage of CTVlowKi-67+ cells in the gate of CD4+ or CD8+ T cells by flow cytometry. Data are presented as percent suppression and determined as follows: % suppression = [1 − (% TRESP cell proliferation in the CD25+ cell–reconstituted PBMCs/% TRESP cell proliferation in the CD25+ cell–depleted PBMCs)] × 100. In this assay, we could evaluate suppressive activity of the total CD25+ TREG cell population, not suppressive activity of TREG cells on a per-cell basis. The depletion of TREG cells from PBMCs and suppression assay were described previously (32). The suppression data were also analyzed by the mitotic index on the basis of the number of mitotic events of TRESP cells. The mitotic index was calculated as described previously (52, 53).
Conventional TREG cell suppression assay
CD4+ cells were isolated from PBMCs using CD4 microbeads (Miltenyi Biotec), LS columns (Miltenyi Biotec), and MACS separator (Miltenyi Biotec). The magnetically labeled CD4+ cells were incubated with fluorochrome-conjugated Abs against cell surface markers (CD25, Live/Dead, CD4, and CD127) at room temperature for 15 min. Using BD FACSAria, we sorted CD4+ TREG cells and non-TREG CD4+ cells. Non-TREG CD4+ cells were labeled with 5 μM CTV (CellTrace Violet Cell Proliferation Kit; Invitrogen) and stimulated with TREG Suppression Inspector (130-092-909; Miltenyi Biotec) with or without the addition of CD4+ TREG cells at a 5:1 ratio. After 96 h of culture, proliferation of CD4+ TRESP cells was analyzed by assessing the percentage of CTVlowKi-67+ cells in the gate of CD4+ T cells by flow cytometry. Data are presented as percent suppression and determined as follows: % suppression = [1 − (% TRESP cell proliferation with TREG cells/% TRESP cell proliferation without TREG cells)] × 100.
Peptide stimulation for intracellular cytokine staining
PBMCs were cultured for 6 h at 37°C with OLP mix for SARS-CoV-2 spike protein (1 µg/ml for each peptide; Miltenyi Biotec). In the culture, we also added anti-human CD28 and CD49d mAbs (1 µg/ml; BD Biosciences) to stimulate costimulatory and adhesion proteins, enhancing the detection of cytokine-producing T cells in intracellular cytokine staining (ICS) assays (54, 55). One hour after the initial stimulation, brefeldin A (GolgiPlug; BD Biosciences) and monensin (GolgiStop; BD Biosciences) were added.
Correlation plots
Corrplot package (v0.84) (56) in R (4.0.1) was used to visualize the Spearman rank correlation coefficients (r) and p values between all parameters. Spearman rank two-tailed p values were calculated using rcorr (Hmisc v4.4-2).
Statistical analysis
Statistical analyses were performed in Prism software version 9.0 for MAC (GraphPad, La Jolla, CA). Data did not follow a normal distribution. To compare values between healthy donors and severe and mild patients, we used one-way ANOVA. To compare paired values for each patient between different time periods (1–14 versus 15–28 DPSO), we performed the mixed-effects analysis test. For patients whose blood samples were drawn on different days within a single time period (1–14 or 15–28 DPSO), the two values were averaged. For cross-sectional analyses, the local regression (LOESS) function was used with a confidence level of 95%. We also performed a linear regression analysis with longitudinally obtained data. Correlations between two parameters were evaluated by a nonparametric two-tailed Spearman correlation test with a 95% confidence interval. A p value ≤0.05 was considered significant.
Data availability
The data that support the findings of this study are available from the corresponding author upon reasonable request.
Results
The CD45RA−FOXP3hi activated TREG cell population is expanded in patients with severe COVID-19
First, we examined the frequency of TREG cells using PBMCs obtained from patients with severe and mild COVID-19 using flow cytometry (Table I). CD3+CD4+FOXP3+CD25hiCD127lo cells were defined as TREG cells (Supplemental Fig. 1A). The relative frequency of TREG cells among CD4+ T cells was plotted during the course of COVID-19 up to 46 DPSO (Fig. 1A) and presented longitudinally for each patient (Fig. 1B). In a linear regression analysis, severe patients exhibited a significant increase in the frequency of TREG cells among CD4+ T cells during the period of 1–21 DPSO, but mild patients did not (Fig. 1C). We determined the mean values obtained between 1 and 14 DPSO for the severe (n = 17) and mild (n = 18) patient groups and found that the TREG frequency was significantly reduced in patients with mild COVID-19 compared with healthy donors (Fig. 1D). We also examined changes in TREG frequency during the course of COVID-19. TREG frequency significantly increased in patients with severe COVID-19 15–28 DPSO compared with 1–14 DPSO, but we found no significant change in patients with mild COVID-19 (Fig. 1E). At 15–28 DPSO, patients with severe disease had a significantly higher TREG frequency than patients with mild disease. We also compared the frequencies of total TREG cells in COVID-19 patients with those in healthy donors 15–28 DPSO (Supplemental Fig. 1B). However, we found no significant difference between patients with severe or mild COVID-19 and healthy donors.
TREG cell frequency among CD4+ T cells in patients with severe or mild COVID-19. (A and B) Scatterplots showing the relationship between DPSO and the frequency of total TREG cells among CD4+ T cells. (A) PBMC samples (n = 120) from patients with COVID-19 (n = 47) were analyzed by flow cytometry. The LOESS nonparametric function is outlined as a black line with gray shading representing the 95% confidence interval. (B) PBMC samples (n = 115) from patients with COVID-19 (n = 42) and healthy donors (n = 16) were analyzed by flow cytometry. The TREG cell frequency of longitudinally tracked samples is represented by colored lines for each patient. (C) Correlation between DPSO and the frequency of TREG cells among CD4+ T cells. (D and E) The frequency of TREG cells among CD4+ T cells was analyzed in longitudinally tracked samples (severe, n = 41; mild, n = 45) from 35 individuals (severe, n = 17; mild, n = 18). (D) The frequency of TREG cells was compared between healthy donors and patients with severe or mild COVID-19 1–14 DPSO. (E) The TREG cell frequencies were compared between paired samples at two time points (1–14 and 15–28 DPSO) and between patients with severe or mild COVID-19 15–28 DPSO. (F) Representative flow cytometry plots of CD45RA−FOXP3hi activated TREG cells among total TREG cells in healthy donors and patients with severe or mild COVID-19 1–14 and 15–28 DPSO. (G and H) Scatterplots showing the relationship between DPSO and the frequency of activated TREG cells among CD4+ TREG cells. (G) The LOESS nonparametric function is outlined as a black line with gray shading representing the 95% confidence interval. (H) The activated TREG cell frequency of longitudinally tracked samples is represented by colored lines for each patient. (I) Correlation between DPSO and the frequency of activated TREG cells among CD4+ T cells. (J and K) The frequency of activated TREG cells among CD4+ T cells analyzed in longitudinally tracked samples. (J) The frequency of activated TREG cells was compared between healthy donors and patients with severe or mild COVID-19 1–14 DPSO. (K) The activated TREG cell frequencies were compared between paired samples at two time points (1–14 and 15–28 DPSO) and between patients with severe or mild COVID-19 15–28 DPSO. Data are presented as the mean ± SD. Statistical analysis was performed using one-way ANOVA (D and J) or the mixed-effects analysis (E and K). For patients with two PBMC samples drawn at a single time period (1–14 or 15–28 DPSO), the two values were averaged (D, E, J, and K). *p < 0.05, **p < 0.01. Data are pooled from seven experiments.
TREG cell frequency among CD4+ T cells in patients with severe or mild COVID-19. (A and B) Scatterplots showing the relationship between DPSO and the frequency of total TREG cells among CD4+ T cells. (A) PBMC samples (n = 120) from patients with COVID-19 (n = 47) were analyzed by flow cytometry. The LOESS nonparametric function is outlined as a black line with gray shading representing the 95% confidence interval. (B) PBMC samples (n = 115) from patients with COVID-19 (n = 42) and healthy donors (n = 16) were analyzed by flow cytometry. The TREG cell frequency of longitudinally tracked samples is represented by colored lines for each patient. (C) Correlation between DPSO and the frequency of TREG cells among CD4+ T cells. (D and E) The frequency of TREG cells among CD4+ T cells was analyzed in longitudinally tracked samples (severe, n = 41; mild, n = 45) from 35 individuals (severe, n = 17; mild, n = 18). (D) The frequency of TREG cells was compared between healthy donors and patients with severe or mild COVID-19 1–14 DPSO. (E) The TREG cell frequencies were compared between paired samples at two time points (1–14 and 15–28 DPSO) and between patients with severe or mild COVID-19 15–28 DPSO. (F) Representative flow cytometry plots of CD45RA−FOXP3hi activated TREG cells among total TREG cells in healthy donors and patients with severe or mild COVID-19 1–14 and 15–28 DPSO. (G and H) Scatterplots showing the relationship between DPSO and the frequency of activated TREG cells among CD4+ TREG cells. (G) The LOESS nonparametric function is outlined as a black line with gray shading representing the 95% confidence interval. (H) The activated TREG cell frequency of longitudinally tracked samples is represented by colored lines for each patient. (I) Correlation between DPSO and the frequency of activated TREG cells among CD4+ T cells. (J and K) The frequency of activated TREG cells among CD4+ T cells analyzed in longitudinally tracked samples. (J) The frequency of activated TREG cells was compared between healthy donors and patients with severe or mild COVID-19 1–14 DPSO. (K) The activated TREG cell frequencies were compared between paired samples at two time points (1–14 and 15–28 DPSO) and between patients with severe or mild COVID-19 15–28 DPSO. Data are presented as the mean ± SD. Statistical analysis was performed using one-way ANOVA (D and J) or the mixed-effects analysis (E and K). For patients with two PBMC samples drawn at a single time period (1–14 or 15–28 DPSO), the two values were averaged (D, E, J, and K). *p < 0.05, **p < 0.01. Data are pooled from seven experiments.
We examined the frequency of CD45RA−FOXP3hi activated TREG cells among CD3+CD4+FOXP3+CD25hiCD127lo TREG cells (Supplemental Fig. 1A). Representative flow cytometry plots from healthy donors and patients with mild and severe COVID-19 are presented in Fig. 1F. The relative frequency of CD45RA−FOXP3hi activated TREG cells among CD4+ T cells was plotted during the course of COVID-19 (Fig. 1G) and presented longitudinally for each patient (Fig. 1H). In a linear regression analysis, severe patients exhibited a significant increase in the frequency of activated TREG cells among CD4+ T cells during the period of 1–21 DPSO, but mild patients did not (Fig. 1I). At 1–14 DPSO, the activated TREG cell frequency was significantly reduced in patients with mild COVID-19 compared with patients with severe COVID-19 (Fig. 1J). The frequency of activated TREG cells significantly increased in patients with severe COVID-19 15–28 DPSO compared with 1–14 DPSO (Fig. 1K). At 15–28 DPSO, patients with severe disease had a significantly higher frequency of activated TREG cells than patients with mild disease. We also compared the frequencies of activated TREG cells in COVID-19 patients with those in healthy donors 15–28 DPSO (Supplemental Fig. 1C). Patients with severe COVID-19 exhibited significantly higher values than healthy donors or patients with mild COVID-19.
We also analyzed the percentage of activated TREG cells among total TREG cells and found that the activated TREG cell percentage was significantly increased in patients with severe COVID-19 compared with patients with mild COVID-19 (Supplemental Fig. 1D). In addition, at 1–14 and 15–28 DPSO, patients with severe disease had a significantly higher percentage of activated TREG cells among total TREG cells than patients with mild disease (Supplemental Fig. 1E). Thus, expansion of the CD45RA−FOXP3hi activated TREG cell population was a characteristic feature of severe COVID-19.
We obtained the absolute numbers of lymphocytes (Supplemental Fig. 1F), total CD3+ (Supplemental Fig. 1G) and CD4+ cells (Supplemental Fig. 1H), TREG cells (Supplemental Fig. 1I), and activated TREG cells (Supplemental Fig. 1J) and compared the numbers between the severe and mild patient groups. As reported previously, the absolute numbers of total lymphocytes and CD3+ and CD4+ cells were significantly lower in the severe group than in the mild group (4, 57). The absolute numbers of total lymphocytes and CD3+ and CD4+ cells were significantly increased 15–28 DPSO only in the mild group. The absolute number of TREG cells did not differ between the two patient groups, but it significantly increased in each group 15–28 DPSO. Importantly, the absolute number of activated TREG cells was significantly higher in severe COVID-19 patients compared with mild COVID-19 patients and significantly increased 15–28 DPSO only in the severe group.
We also analyzed the normalized geometric mean fluorescence intensity of FOXP3 in each sample. There was no significant difference in the normalized geometric mean fluorescence intensity of FOXP3 in TREG cells among healthy controls and patients with severe and mild COVID-19, although it tended to be higher in patients with severe disease (Supplemental Fig. 1K, 1L).
Single-cell transcriptomic analysis reveals proliferating, but apoptosis-prone, features in TREG cells from patients with severe COVID-19
To comprehensively understand the molecular characteristics of TREG cells in patients with COVID-19, we reanalyzed publicly available single-cell RNA sequencing (scRNA-seq) data for SARS-CoV-2–reactive CD4+ T cells (GSE152522; https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE152522) (47). In the previous study, SARS-CoV-2–reactive CD4+ T cells had been enriched based on the upregulation of activation-induced markers, such as CD137 and CD69, after ex vivo stimulation of PBMCs with SARS-CoV-2 Ags, such as membrane and spike. The enriched SARS-CoV-2–reactive CD4+ T cells from patients with COVID-19 (mild, n = 21; severe, n = 9) had been subjected to scRNA-seq analysis. In this study, we reanalyzed the raw count matrices and metadata. The MNN algorithm was used to correct the batch effect according to donor origin (49). We clustered the total SARS-CoV-2–reactive CD4+ T cells using the UMAP algorithm with the Seurat package (48). TREG cells with FOXP3 expression clustered among CD4+ T cells (Supplemental Fig. 2A, 2B). TREG cells were further subclustered into four different populations (Fig. 2A). These subclusters were characterized according to the expression of differentially expressed marker genes (Fig. 2B, Supplemental Fig. 2C, 2D).
Analysis of the scRNA-seq data for SARS-CoV-2–reactive CD4+ TREG cells from COVID-19 patients. Publicly available scRNA-seq data for SARS-CoV-2–reactive CD4+ T cells (GSE152522) (47) were reanalyzed. The enriched SARS-CoV-2–reactive CD4+ T cells from patients with COVID-19 (mild, n = 21; severe, n = 9) had been subjected to scRNA-seq analysis. (A) UMAP depicting clusters of SARS-CoV-2–reactive TREG cells with FOXP3 expression. (B) Dot plot showing the representative marker genes of each cluster. (C) Bar plots showing the −log p value from the GSEA of immune-related GO biological pathway terms in DEGs from each cluster. (D) Dot plot showing the module scores of gene sets related to cell cycle, S, and G2M phase among each cluster. (E) Proportion of each cluster according to disease severity. (F) Dot plot showing gene expression and module scores for gene sets related to apoptosis, including TP53 and the regulation of p53- and ER stress–mediated apoptosis. Statistical analysis was performed using the Wilcoxon rank sum t test for DEGs and unpaired nonparametric Mann–Whitney t test for comparing proportions. *p < 0.05, **p < 0.01.
Analysis of the scRNA-seq data for SARS-CoV-2–reactive CD4+ TREG cells from COVID-19 patients. Publicly available scRNA-seq data for SARS-CoV-2–reactive CD4+ T cells (GSE152522) (47) were reanalyzed. The enriched SARS-CoV-2–reactive CD4+ T cells from patients with COVID-19 (mild, n = 21; severe, n = 9) had been subjected to scRNA-seq analysis. (A) UMAP depicting clusters of SARS-CoV-2–reactive TREG cells with FOXP3 expression. (B) Dot plot showing the representative marker genes of each cluster. (C) Bar plots showing the −log p value from the GSEA of immune-related GO biological pathway terms in DEGs from each cluster. (D) Dot plot showing the module scores of gene sets related to cell cycle, S, and G2M phase among each cluster. (E) Proportion of each cluster according to disease severity. (F) Dot plot showing gene expression and module scores for gene sets related to apoptosis, including TP53 and the regulation of p53- and ER stress–mediated apoptosis. Statistical analysis was performed using the Wilcoxon rank sum t test for DEGs and unpaired nonparametric Mann–Whitney t test for comparing proportions. *p < 0.05, **p < 0.01.
To further investigate the molecular characteristics of each cluster, we performed GSEA with publicly available gene sets related to immune responses (Fig. 2C). Cluster-1 was characterized by gene sets related to the response to type I IFN and the IFN-γ and cytokine response. Cluster-2 was mainly enriched with gene sets related to apoptosis, especially the intrinsic apoptotic pathway. In further analyses, cluster-2 was also enriched with gene sets related to S phase of the cell cycle (Fig. 2D). Collectively, cluster-2 exhibits proliferating but apoptosis-prone features, indicating that TREG cells in cluster-2 are recently activated but vulnerable to apoptotic stimulation. Cluster-3 was characterized by genes related to leukocyte chemotaxis, and cluster-4 was characterized by marked upregulation of CCR8 in addition to gene sets related to the regulation of diverse immune effector processes.
Next, we compared the proportion of each cluster according to disease severity. Cluster-2 was significantly increased in patients with severe COVID-19 (mean 54.2%) compared with patients with mild COVID-19 (mean 38.4%), whereas cluster-4 was significantly decreased in patients with severe disease (mean 1.8%) compared with patients with mild disease (mean 5.4%; Fig. 2E). The significant increase in the proportion of cluster-2 in patients with severe COVID-19 indicates that TREG cells are activated during severe COVID-19 but become vulnerable to apoptosis. To further analyze the enrichment of gene sets related to apoptosis, we identified TREG cells with active gene sets using the AUCell method (51). When we plotted cells with a gene set enrichment score above the threshold, cells with various apoptotic features were mainly located in cluster-2 (Supplemental Fig. 2E). In addition, the expression of various apoptosis-related gene sets was enriched in patients with severe disease compared with patients with mild disease (Fig. 2F). Collectively, these findings demonstrate that TREG cells from patients with severe COVID-19 exhibit proliferating but apoptosis-prone features.
TREG cells from patients with severe COVID-19 exhibit enhanced apoptosis
We validated the results of the scRNA-seq analysis using PBMCs obtained from our cohort. First, we examined the expression of Ki-67, a marker of proliferating cells, among TREG cells (Fig. 3A). The relative frequency of Ki-67+ cells among TREG cells was plotted during the course of COVID-19 (Fig. 3B) and presented longitudinally for each patient (Fig. 3C). In a linear regression analysis, severe patients exhibited a significant increase in the frequency of Ki-67+ cells among TREG cells during the period of 1–21 DPSO, but mild patients did not (Fig. 3D). At 1–14 DPSO, the frequency of Ki-67+ cells among TREG cells showed no significant differences between healthy donors and patients with COVID-19 (Fig. 3E). Although the frequency of Ki-67+ cells among TREG cells was not increased in patients with severe COVID-19 1–14 DPSO, it significantly increased in patients with severe COVID-19 at 15–28 DPSO compared with 1–14 DPSO, but there was no significant change in patients with mild COVID-19 (Fig. 3F). At 15–28 DPSO, patients with severe disease had a significantly higher frequency of Ki-67+ cells than patients with mild disease. We also compared the frequencies of Ki-67+ TREG cells in COVID-19 patients with those in healthy donors 15–28 DPSO (Fig. 3G). Patients with mild COVID-19 exhibited significantly lower values than patients with severe COVID-19. However, we found no difference between patients with severe COVID-19 and healthy donors. Thus, TREG cells undergo proliferation in patients with severe COVID-19, corroborating the results from the scRNA-seq analysis.
Proliferation and apoptotic features of TREG cells. (A) Representative flow cytometry plots of Ki-67+ cells among TREG cells from healthy donors and patients with severe or mild COVID-19 1–14 and 15–28 DPSO. (B and C) Scatterplots showing the relationship between DPSO and the frequency of Ki-67+ cells among TREG cells. (B) PBMC samples (n = 120) from patients with COVID-19 (n = 47) were analyzed by flow cytometry. The LOESS nonparametric function is outlined as a black line with gray shading representing the 95% confidence interval. (C) PBMC samples (n = 115) from patients with COVID-19 (n = 42) and healthy donors (n = 16) were analyzed by flow cytometry. The Ki-67+ TREG cell frequency of longitudinally tracked samples is represented by colored lines for each patient. (D) Correlation between DPSO and the frequency of Ki-67+ cells among TREG cells. (E–G) The frequency of Ki-67+ cells among TREG cells was analyzed in longitudinally tracked samples (severe, n = 41; mild, n = 45) from 35 individuals (severe, n = 17; mild, n = 18). (E) The frequency of Ki-67+ cells among TREG cells was compared between healthy donors and patients with severe or mild COVID-19 1–14 DPSO. (F) Ki-67+ TREG cell frequencies were compared between paired samples at two time points (1–14 and 15–28 DPSO) and between patients with severe or mild COVID-19 15–28 DPSO. (G) The frequency of Ki-67+ cells among TREG cells was compared between healthy donors and patients with severe or mild COVID-19 15–28 DPSO. (H–K) PBMC samples (n = 24) from patients with severe (n = 12) or mild (n = 8) COVID-19 were stimulated by anti-CD3/CD28 Abs for 12 h. Representative flow cytometry plots (H) and the frequency of annexin V+Live/Dead dye+ cells among TREG cells (I) in patients with severe or mild COVID-19. Representative flow cytometry plots (J) and the frequency of active capase-3+ cells among TREG cells (K) in patients with severe or mild COVID-19. Data are presented as the mean ± SD. Statistical analysis was performed using one-way ANOVA (E and G) or the mixed-effects analysis (F, I, and K). For patients with two PBMC samples drawn at a single time period (1–14 or 15–28 DPSO), the two values were averaged (E–G). *p < 0.05, **p < 0.01, ****p < 0.0001. Data are pooled from seven (A–G) or four (H–K) experiments.
Proliferation and apoptotic features of TREG cells. (A) Representative flow cytometry plots of Ki-67+ cells among TREG cells from healthy donors and patients with severe or mild COVID-19 1–14 and 15–28 DPSO. (B and C) Scatterplots showing the relationship between DPSO and the frequency of Ki-67+ cells among TREG cells. (B) PBMC samples (n = 120) from patients with COVID-19 (n = 47) were analyzed by flow cytometry. The LOESS nonparametric function is outlined as a black line with gray shading representing the 95% confidence interval. (C) PBMC samples (n = 115) from patients with COVID-19 (n = 42) and healthy donors (n = 16) were analyzed by flow cytometry. The Ki-67+ TREG cell frequency of longitudinally tracked samples is represented by colored lines for each patient. (D) Correlation between DPSO and the frequency of Ki-67+ cells among TREG cells. (E–G) The frequency of Ki-67+ cells among TREG cells was analyzed in longitudinally tracked samples (severe, n = 41; mild, n = 45) from 35 individuals (severe, n = 17; mild, n = 18). (E) The frequency of Ki-67+ cells among TREG cells was compared between healthy donors and patients with severe or mild COVID-19 1–14 DPSO. (F) Ki-67+ TREG cell frequencies were compared between paired samples at two time points (1–14 and 15–28 DPSO) and between patients with severe or mild COVID-19 15–28 DPSO. (G) The frequency of Ki-67+ cells among TREG cells was compared between healthy donors and patients with severe or mild COVID-19 15–28 DPSO. (H–K) PBMC samples (n = 24) from patients with severe (n = 12) or mild (n = 8) COVID-19 were stimulated by anti-CD3/CD28 Abs for 12 h. Representative flow cytometry plots (H) and the frequency of annexin V+Live/Dead dye+ cells among TREG cells (I) in patients with severe or mild COVID-19. Representative flow cytometry plots (J) and the frequency of active capase-3+ cells among TREG cells (K) in patients with severe or mild COVID-19. Data are presented as the mean ± SD. Statistical analysis was performed using one-way ANOVA (E and G) or the mixed-effects analysis (F, I, and K). For patients with two PBMC samples drawn at a single time period (1–14 or 15–28 DPSO), the two values were averaged (E–G). *p < 0.05, **p < 0.01, ****p < 0.0001. Data are pooled from seven (A–G) or four (H–K) experiments.
We also evaluated apoptosis in TREG cells from COVID-19 patients with or without anti-CD3/anti-CD28 stimulation, which mimics TCR-mediated stimulation using PBMCs obtained 15–28 DPSO. When apoptotic cells were detected by staining with annexin V and Live/Dead dye, the frequency of apoptotic cells among TREG cells was significantly increased by anti-CD3/anti-CD28 stimulation in patients with severe disease, but not in patients with mild disease (Fig. 3H, 3I). The frequency of apoptotic cells after stimulation was significantly higher in patients with severe disease than in patients with mild disease. Similar results were observed when apoptotic cells were detected by immunostaining active, cleaved caspase-3 (Fig. 3J, 3K). These findings confirmed the results from scRNA-seq analysis that TREG cells from patients with severe COVID-19 are prone to apoptosis. Taken together, TREG cells from patients with severe COVID-19 easily undergo apoptosis upon stimulation, although they have features of activation and proliferation.
TREG suppressive functions are comparable among healthy donors and patients with severe and mild COVID-19
Because the TREG cell population underwent dynamic changes in patients with severe COVID-19, including an increased frequency of activated TREG cells and enhanced apoptotic features, we examined the suppressive functions of the TREG population. First, we investigated the expression of CTLA-4 (Fig. 4A), which is a main mediator of the suppressive functions of TREG cells (20–22). The relative frequency of CTLA-4+ cells among TREG cells was plotted during the course of COVID-19 (Fig. 4B) and presented longitudinally for each patient (Fig. 4C). In a linear regression analysis, severe patients tended to have an increased frequency of CTLA-4+ cells among TREG cells during the period of 1–21 DPSO, but mild patients did not (Fig. 4D). The frequency of CTLA-4+ cells significantly increased 1–14 DPSO in patients with severe COVID-19 compared with healthy donors and patients with mild COVID-19 (Fig. 4E), supporting a previous report that genes encoding suppressive molecules, such as CTLA-4, are up-regulated in TREG cells from patients with severe COVID-19 (44). At 1–14 and 15–28 DPSO, patients with severe disease had a significantly higher frequency of CTLA-4+ cells than patients with mild disease (Fig. 4F). The absolute number of CTLA-4+ TREG cells was significantly higher in the severe group compared with the mild group at 15–28 DPSO and significantly increased at 15–28 DPSO only in the severe group (Fig. 4G).
CTLA-4 expression and suppressive activity of TREG cells. (A) Representative flow cytometry plots of CTLA-4+ cells among TREG cells in healthy donors and patients with severe or mild COVID-19 at 1–14 and 15–28 DPSO. (B and C) Scatterplots showing the relationship between DPSO and the frequency of CTLA-4+ cells among TREG cells. (B) PBMC samples (n = 120) from patients with COVID-19 (n = 47) were analyzed by flow cytometry. The LOESS nonparametric function is outlined as a black line with gray shading, representing the 95% confidence interval. (C) PBMC samples (n = 115) from patients with COVID-19 (n = 42) and healthy donors (n = 16) were analyzed by flow cytometry. The CTLA-4+ TREG cell frequency of longitudinally tracked samples is represented by colored lines for each patient. (D) Correlation between DPSO and the frequency of CTLA-4+ cells among TREG cells. (E and F) The frequency of CTLA-4+ cells among TREG cells was analyzed in longitudinally tracked samples (severe, n = 41; mild, n = 45) from 35 individuals (severe, n = 17; mild, n = 18). (E) The frequency of CTLA-4+ cells among TREG cells was compared between healthy donors and patients with severe or mild COVID-19 1–14 DPSO. (F) CTLA-4+ TREG cell frequencies were compared between paired samples at two time points (1–14 and 15–28 DPSO) and between patients with severe or mild COVID-19 15–28 DPSO. (G) The absolute counts of CTLA-4+ TREG cells were compared between paired samples at two time points (1–14 and 15–28 DPSO) and between patients with severe or mild COVID-19 at the noted time points. (H–J) The suppressive activity of CD25+ TREG cells in PBMC samples (healthy donors, n = 7; severe, n = 8; mild, n = 8) was analyzed by the proliferation of CD25+ cell–depleted PBMCs and CD25+ cell–reconstituted PBMCs. (H) Representative flow cytometry plots of CTVlo cells among CD4+CD25− or CD8+ Tresp cells with or without CD25+ TREG cells. The proportion of suppression of CD4+CD25− T cells (I) and CD8+ T cells (J) was compared in healthy donors and patients with severe or mild COVID-19. Mean percentages are represented by horizontal bars. Data are presented as the mean ± SD. Statistical analysis was performed using one-way ANOVA (E, I, and J) or the mixed-effects analysis (F and G). For patients with two PBMC samples drawn at a single time period (1–14 or 15–28 DPSO), the two values were averaged (E–G). **p < 0.01, ***p < 0.001. Data are pooled from seven (A–G) or three (H–J) experiments.
CTLA-4 expression and suppressive activity of TREG cells. (A) Representative flow cytometry plots of CTLA-4+ cells among TREG cells in healthy donors and patients with severe or mild COVID-19 at 1–14 and 15–28 DPSO. (B and C) Scatterplots showing the relationship between DPSO and the frequency of CTLA-4+ cells among TREG cells. (B) PBMC samples (n = 120) from patients with COVID-19 (n = 47) were analyzed by flow cytometry. The LOESS nonparametric function is outlined as a black line with gray shading, representing the 95% confidence interval. (C) PBMC samples (n = 115) from patients with COVID-19 (n = 42) and healthy donors (n = 16) were analyzed by flow cytometry. The CTLA-4+ TREG cell frequency of longitudinally tracked samples is represented by colored lines for each patient. (D) Correlation between DPSO and the frequency of CTLA-4+ cells among TREG cells. (E and F) The frequency of CTLA-4+ cells among TREG cells was analyzed in longitudinally tracked samples (severe, n = 41; mild, n = 45) from 35 individuals (severe, n = 17; mild, n = 18). (E) The frequency of CTLA-4+ cells among TREG cells was compared between healthy donors and patients with severe or mild COVID-19 1–14 DPSO. (F) CTLA-4+ TREG cell frequencies were compared between paired samples at two time points (1–14 and 15–28 DPSO) and between patients with severe or mild COVID-19 15–28 DPSO. (G) The absolute counts of CTLA-4+ TREG cells were compared between paired samples at two time points (1–14 and 15–28 DPSO) and between patients with severe or mild COVID-19 at the noted time points. (H–J) The suppressive activity of CD25+ TREG cells in PBMC samples (healthy donors, n = 7; severe, n = 8; mild, n = 8) was analyzed by the proliferation of CD25+ cell–depleted PBMCs and CD25+ cell–reconstituted PBMCs. (H) Representative flow cytometry plots of CTVlo cells among CD4+CD25− or CD8+ Tresp cells with or without CD25+ TREG cells. The proportion of suppression of CD4+CD25− T cells (I) and CD8+ T cells (J) was compared in healthy donors and patients with severe or mild COVID-19. Mean percentages are represented by horizontal bars. Data are presented as the mean ± SD. Statistical analysis was performed using one-way ANOVA (E, I, and J) or the mixed-effects analysis (F and G). For patients with two PBMC samples drawn at a single time period (1–14 or 15–28 DPSO), the two values were averaged (E–G). **p < 0.01, ***p < 0.001. Data are pooled from seven (A–G) or three (H–J) experiments.
Next, we evaluated the suppressive activity of the TREG population by assessing the anti-CD3/CD28–stimulated proliferation of TREG-depleted PBMCs and TREG-reconstituted PBMCs. The proliferation of non-TREG CD4+ and CD8+ T cells was measured by CTV dilution (Fig. 4H). In this assay, we evaluated the suppressive activity of the total TREG population, not the suppressive activity of TREG cells on a per-cell basis. We found that the suppressive activity of the TREG population was not significantly different among healthy donors and patients with severe or mild COVID-19 when the proliferation of non-TREG CD4+ T cells (Fig. 4I) and CD8+ T cells (Fig. 4J) was assessed. Similar results were obtained when we analyzed the suppression data by the mitotic index on the basis of the number of mitotic events of TRESP cells (Supplemental Fig. 3A, 3B).
We also performed conventional TREG cell suppression assays on a per-cell basis using sorted TREG cells and non-TREG CD4+ T cells and found that the suppressive activity of TREG cells was not significantly different among healthy donors and patients with severe or mild COVID-19 (Supplemental Fig. 3C).
Collectively, these results demonstrate that the suppressive functions of the TREG cell population are not enhanced in patients with severe COVID-19, although the frequencies of activated TREG cells and CTLA-4+ TREG cells are increased in patients with severe COVID-19. This finding can be explained by increased apoptosis of TREG cells in patients with severe COVID-19. The TREG cell population is remarkably deranged during severe COVID-19, but the overall suppressive activity of the TREG cell population is not altered.
TREG cell frequency inversely correlates with CD4+ T cell effector functions in patients with mild COVID-19, but not severe COVID-19
ICS was performed to assess the SARS-CoV-2–specific effector functions of CD4+ and CD8+ T cells. In ICS, PBMCs were ex vivo stimulated with OLP mix for SARS-CoV-2 spike protein and stained for IFN-γ, IL-2, and TNF (Supplemental Fig. 3D). The frequency of IFN-γ– or TNF-producing T cells was not different between patients with severe and mild COVID-19, whereas the frequency of IL-2–producing T cells was significantly higher in patients with severe COVID-19 (Supplemental Fig. 3E, 3F). The absolute count of TNF-producing T cells was significantly higher in patients with mild COVID-19 (Supplemental Fig. 3G, 3H). The ratio of IFN-γ–producing CD4+ T cells or TNF-producing CD8+ T cells to TREG cells was significantly higher in patients with mild COVID-19 (Supplemental Fig. 3I, 3J).
We analyzed whether the frequency of TREG cells correlates with immunological parameters. We analyzed 21 parameters, including demographic, leukocyte, TREG, CD4 effector function, and CD8 effector function parameters, in correlograms. The absolute number of TREG cells positively correlated with the absolute number of CD3+, CD4+, and CD8+ T cells in both mild and severe patients. However, the absolute number of neutrophils and the SARS-CoV-2 neutralizing Ab titers did not significantly correlate with the TREG cell population regardless of the severity of the disease. In patients with mild COVID-19, the TREG-related parameters exhibited significant inverse correlations with CD4 T cell effector functions (blue box in Fig. 5A). However, such correlations were not observed in patients with severe COVID-19 (blue box in Fig. 5B).
Association of TREG cell frequency with immunological parameters. Frequency of TREG and activated TREG cells was analyzed using PBMC samples (n = 120) from patients with COVID-19 (n = 47) by flow cytometry. Frequency of SARS-CoV-2 spike-specific, cytokine-producing CD4+ and CD8+ T cells was analyzed using PBMC samples (≥15 DPSO) from patients with severe (n = 16) or mild (n = 23) COVID-19 by ICS. In ICS, PBMCs were ex vivo stimulated with OLP mix for SARS-CoV-2 spike protein (1 µg/ml for each peptide) for 6 h and stained for IFN-γ, IL-2, and TNF. The frequency of polyfunctional T cells that produce ≥2 cytokines simultaneously was also determined. (A and B) Correlogram of patients with mild (A) or severe (B) COVID-19. Correlation values (R) were analyzed using Spearman rank and are shown in red (1.0) to blue (−1.0). Statistical analyses were performed as pairwise correlations. Squares indicate a significant correlation. (C, E, and G) Correlation between the frequency of IFN-γ+, IL-2+, or TNF+ cells among CD4+ T cells and the frequency of TREG cells among CD4+ T cells in total (C), mild (E), and severe (G) patients. (D, F, and H) Correlation between the frequency of polyfunctional cells among CD4+ T cells and the frequency of TREG cells among CD4+ T cells in total (C), mild (E), and severe (G) patients. aTREG, activated TREG. *p < 0.05, **p < 0.01, ***p < 0.001. Data are pooled from four experiments.
Association of TREG cell frequency with immunological parameters. Frequency of TREG and activated TREG cells was analyzed using PBMC samples (n = 120) from patients with COVID-19 (n = 47) by flow cytometry. Frequency of SARS-CoV-2 spike-specific, cytokine-producing CD4+ and CD8+ T cells was analyzed using PBMC samples (≥15 DPSO) from patients with severe (n = 16) or mild (n = 23) COVID-19 by ICS. In ICS, PBMCs were ex vivo stimulated with OLP mix for SARS-CoV-2 spike protein (1 µg/ml for each peptide) for 6 h and stained for IFN-γ, IL-2, and TNF. The frequency of polyfunctional T cells that produce ≥2 cytokines simultaneously was also determined. (A and B) Correlogram of patients with mild (A) or severe (B) COVID-19. Correlation values (R) were analyzed using Spearman rank and are shown in red (1.0) to blue (−1.0). Statistical analyses were performed as pairwise correlations. Squares indicate a significant correlation. (C, E, and G) Correlation between the frequency of IFN-γ+, IL-2+, or TNF+ cells among CD4+ T cells and the frequency of TREG cells among CD4+ T cells in total (C), mild (E), and severe (G) patients. (D, F, and H) Correlation between the frequency of polyfunctional cells among CD4+ T cells and the frequency of TREG cells among CD4+ T cells in total (C), mild (E), and severe (G) patients. aTREG, activated TREG. *p < 0.05, **p < 0.01, ***p < 0.001. Data are pooled from four experiments.
When regression analyses were performed between TREG cell frequency and CD4 T cell effector functions among all patients with COVID-19, no significant correlation was observed (Fig. 5C, 5D). In patients with mild COVID-19, the frequency of TREG cells among CD4+ T cells inversely correlated with the frequency of spike-specific CD4+ T cells that produced effector cytokines, including IFN-γ, IL-2, and TNF, among CD4+ T cells (Fig. 5E). We also analyzed the frequency of polyfunctional CD4+ T cells that produce ≥2 cytokines simultaneously. We found that the frequency of TREG cells among CD4+ T cells inversely correlated with the frequency of spike-specific polyfunctional CD4+ T cells in patients with mild COVID-19 (Fig. 5F). However, such correlations were not observed in patients with severe COVID-19 (Fig. 5G, 5H). These findings suggest that TREG cells negatively control the SARS-CoV-2–specific effector functions of CD4+ T cells during mild COVID-19. However, such negative correlations were not observed in patients with severe COVID-19. The lack of correlation might be because of functional derangement of the TREG cell population during severe COVID-19 (Table II).
. | Mild COVID-19 . | Severe COVID-19 . |
---|---|---|
Increase in total TREG cell frequency | − | + |
Increase in total activated TREG cell frequency | − | + |
Increase in Ki-67+ proliferating TREG cell frequency | − | + |
Increase in CTLA-4+ TREG cell frequency | − | + |
Apoptosis of TREG cells | < | |
Suppressive activity of the TREG cell population | ≈ | |
Correlation between TREG cell frequency and SARS-CoV-2–specific CD4+ T cell functions | Inverse correlation | No correlation |
. | Mild COVID-19 . | Severe COVID-19 . |
---|---|---|
Increase in total TREG cell frequency | − | + |
Increase in total activated TREG cell frequency | − | + |
Increase in Ki-67+ proliferating TREG cell frequency | − | + |
Increase in CTLA-4+ TREG cell frequency | − | + |
Apoptosis of TREG cells | < | |
Suppressive activity of the TREG cell population | ≈ | |
Correlation between TREG cell frequency and SARS-CoV-2–specific CD4+ T cell functions | Inverse correlation | No correlation |
Discussion
This longitudinal study demonstrated the phenotypes and functions of the CD4+CD25+FOXP3+ TREG cell population in patients with severe and mild COVID-19 as summarized in Table II. In patients with severe COVID-19, the frequencies of total TREG cells and CD45RA−FOXP3hi activated TREG cells among CD4+ T cells significantly increased 15–28 DPSO. At this time point, TREG cells from severe patients showed not only increased proliferation but also enhanced apoptotic features. Although the expression of CTLA-4, an effector molecule for immunosuppressive functions, significantly increased 15–28 DPSO in TREG cells from severe patients, the actual suppressive function of the TREG cell population in severe patients was comparable with that of healthy donors or mild patients. Interestingly, the frequency of TREG cells among CD4+ T cells inversely correlated with SARS-CoV-2–specific cytokine production and the polyfunctionality of CD4+ T cells in mild patients, but not severe patients. These data suggest that TREG cells are major regulators of virus-specific CD4+ T cell responses during mild COVID-19, whereas the TREG cell population is functionally deranged during severe COVID-19. Our findings suggest that the TREG cell population alters distinctively in patients with severe and mild COVID-19 during the course of disease.
Several studies have examined the landscape of immune cell populations in patients with COVID-19 by performing transcriptomic analyses and reported alteration in the TREG cell population according to the severity of the disease (43, 44, 58). Recent studies showed consistent results that the proportion of TREG cells and their FOXP3 expression level increased in patients with severe COVID-19, corroborating our current finding (43, 44). In addition, transcriptomic analysis showed proinflammatory features and suppressive features in TREG cells from patients with severe COVID-19 (43). They suggest that IL-6 and IL-18 might cause the perturbation in the TREG cell population. Another study reported that TREG cells from patients with COVID-19 exhibit activated signatures, which increases with the disease severity (44). Relatively decreased frequencies of TREG cells were reported in convalescent patients after mild COVID-19 (45). However, previous studies have not directly investigated the suppressive capacity of TREG cells and the relationship between TREG cells and SARS-CoV-2–specific effector T cell functions. In addition, previous studies examined TREG cells only cross-sectionally and lacked longitudinal analysis of each COVID-19 patient.
In this study, we reanalyzed publicly available scRNA-seq data for SARS-CoV-2–reactive CD4+ T cells that had been enriched based on the upregulation of activation-induced markers CD137 and CD69 after ex vivo stimulation of PBMCs with SARS-CoV-2 Ags (47). TREG cells from patients with severe COVID-19 exhibited apoptosis-prone features. However, ex vivo stimulation with SARS-CoV-2 Ags may alter the transcriptomic features of TREG cells. To overcome this limitation, we performed additional scRNA-seq reanalysis using another set of scRNA-seq data from patients with COVID-19 (59) and confirmed the apoptosis-prone features of TREG cells from patients with severe COVID-19.
Previous studies regarding the dynamics of the TREG cell population during viral infection have shown that increased TREG cell frequencies or suppressive activity may be disadvantageous to the host in regard to eliminating infecting viruses (34, 37, 38). The frequencies of TREG cells in the PB and liver have been reported to be higher in patients with HCV infection (29–31). Attenuated CD8+ T cell functions are related to increased TREG cell frequency during HCV infection. In addition, therapeutic vaccine-induced enhancement of HCV-specific T cell responses is associated with an IFNL3 adjuvant-related reduction in TREG cell frequency in patients with chronic HCV infection (60), indicating that TREG cells are continuously suppressing virus-specific T cell responses during chronic HCV infection. In this study, the frequency of TREG cells was lower in patients with mild COVID-19 than in healthy donors or severe patients, and these cells exhibited less activated phenotypes. Moreover, the frequency of TREG cells inversely correlated with SARS-CoV-2–specific cytokine production and the polyfunctionality of CD4+ T cells in mild COVID-19 patients. Our study supports that the TREG cell population from mild COVID-19 patients is not abnormally disturbed and able to regulate the effector functions of virus-specific effector T cells.
Our study has limitations. First, patients with severe and mild COVID-19 differed in their age distribution (average age 62.9 and 47.8 y in severe and mild patients, respectively). In addition, the two patient groups differed in their gender distribution. Further studies of TREG cells with age- and sex-matched patient groups are required. Second, we could not examine whether our current findings are generalizable to patients with other severe respiratory viral diseases, such as influenza and respiratory syncytial virus infection.
In this study, the size of the TREG cell population did not correlate with the effector functions of virus-specific CD4+ T cells during severe COVID-19, showing that the TREG cell population is aberrantly deranged. The TREG cell population exhibited enhanced apoptosis in patients with severe COVID-19. Moreover, the scRNA-seq analysis of public data revealed that TREG cells are prone to intrinsic pathway-mediated apoptosis. A previous study demonstrated that the frequency of TREG cells is decreased because of upregulation of Fas receptor during acute HAV infection (32). Importantly, a decreased frequency of TREG cells was significantly associated with enhanced liver injury in patients with acute HAV infection. Given that the liver injury is caused by a T cell–mediated immunopathological mechanism during acute HAV infection (33), enhanced apoptosis of TREG cells results in a loss of immunosuppression and exaggerated immunopathological host injury in patients with acute HAV infection. Other studies support the TREG cell population being regulated mainly by susceptibility to Fas-mediated apoptosis (61–63). Although our findings demonstrate increased cleavage of capsase-3, which is a downstream caspase, in the expanded TREG cell population from severe COVID-19 patients, the precise mechanism of how apoptosis is initiated in TREG cells needs to be investigated further.
Immunosuppressive cells other than TREG cells also contribute to the maintenance of immune homeostasis, including myeloid-derived suppressor cells (MDSCs). MDSCs are known to exert their immunosuppressive activity on T cells, macrophages, dendritic cells, and NK cells in pathological environments through reactive oxygen species or l-arginine. In viral infection and cancer, MDSCs have been shown to suppress the functions of other immune effector cells and increase the number of TREG cells (64–67). In patients infected with SARS-CoV-2, increased proinflammatory cytokines in severe COVID-19 patients (16) may cause the expansion of MDSCs, which are capable of inhibiting the proliferation and function of effector T cells (68–70). Severe progression of COVID-19 may be associated with perturbation and derangement of the major two immunosuppressive cell types, including TREG cells and MDSCs.
In this study, we found that the TREG cell population from patients with severe COVID-19 expanded significantly and exhibited not only more proliferative features but also elevated apoptotic features compared with the population from mild patients at 15–28 DPSO. Despite the increased activation, we observed comparable suppressive activity of the TREG cell populations of severe and mild COVID-19 patients. The frequency of TREG cells in mild patients inversely correlated with the functionality of SARS-CoV-2–specific CD4+ T cells, supporting the importance of TREG cells in the regulation of virus-specific CD4+ T cells in mild COVID-19. However, no correlations were observed in patients with severe disease. Our findings suggest that the dynamics and functions of the TREG cell population are distinctive in patients with severe and mild COVID-19.
Disclosures
The authors have no financial conflicts of interest.
Acknowledgments
We appreciate laboratory members for support and critical opinions.
Footnotes
This work was supported by the Institute for Basic Science, Korea, under project code IBS-R801-D2.
The online version of this article contains supplemental material.
- CTV
CellTrace Violet
- DEG
differentially expressed gene
- DPSO
days postsymptom onset
- GSEA
gene set enrichment analysis
- HAV
hepatitis A virus
- HCV
hepatitis C virus
- ICS
intracellular cytokine staining
- LOESS
local regression
- MDSC
myeloid-derived suppressor cell
- MNN
mutual nearest neighbor
- OLP
overlapping peptide
- PB
peripheral blood
- scRNA-seq
single-cell RNA sequencing
- TREG
regulatory T
- TRESP
responder T
- UMAP
uniform manifold approximation and projection