Obstructive sleep apnea syndrome (OSAS) represents a substantial disease of recurrent sleep fragmentation, leading to intermittent hypoxia and subsequent diseases such as cardiovascular, metabolic, or cognitive dysfunctions. In addition, OSAS is considered as low-grade systemic inflammation, which is associated with a higher incidence of cancer, severity of infections, and an overall immune dysregulation. This research project aims to comprehensively investigate the interplay of wholesome sleep and the immune functions of circulating monocytes and T cells in OSAS patients, which are known to be affected by oxidative stress. We studied the distribution of the CD14/CD16 characterized monocyte subsets in peripheral blood as well as their PD-L1 expression and complex formation with T cells. Furthermore, a detailed analysis of T cell subsets with regard to their PD-1 and PD-L1 expression was performed. Data revealed a decrease of classical monocytes accompanied by an increase of both CD16+ monocyte subsets in OSAS patients that was positively correlated with the body mass index. OSAS patients revealed an increased PD-1 and PD-L1 expression in T cells and monocytes, respectively, which was linked to the severity of monocyte subset alterations. The complex formation of monocytes and T cells was also elevated in OSAS patients, which indicates a deregulated PD-1/PD-L1 cross-talk between these cells. Our data show for the first time, to our knowledge, massive alterations of peripheral monocyte subsets in response to OSAS and its accompanying phenomena.

Sleep is of fundamental importance for all living beings, although its manifold contributions to the fundamentals of life are still somewhat of a mystery. One of the vital roles of sleep is to consolidate the incredible amount of information our brains have to handle every day. Furthermore, wholesome sleep is essential to secure metabolite clearance, tissue repair, or hormone biosynthesis (14).

Obstructive sleep apnea syndrome (OSAS), in contrast, represents a substantial disease of recurrent breathing intermissions, resulting in hypopnea/apnea followed by reoxygenation. Up to 9% of the adult population suffer from this potentially life-threatening disorder. Besides reduced quality of life, OSAS is a major risk factor for cardiovascular diseases, metabolic disorders, and cognitive dysfunctions (5). Moreover, wholesome sleep and normal circadian patterns are highly important for a functioning immune system. Because of nocturnal oxidative stress, OSAS is considered a low-grade systemic inflammation that is initiated by intermittent hypoxia and sleep fragmentation that leads to activation of NF-κB and HIF-1α (3, 6). These promote the activation of immune cells, especially lymphocytes and monocytes that secrete high amounts of inflammatory cytokines and adhesion molecules (6, 7). Several studies demonstrated increased levels of C-reactive protein, TNF-α, IL-6, IL-8, ICAM, and VCAM in OSAS patients (6, 8, 9). Consequently, excessive adherence of immune cells and an overall prevailing proinflammatory milieu leads to vascular injury and the development of atherosclerosis and cardiovascular diseases (1012).

OSAS-induced hypoxic stress increases the invasion of monocytes into the endothelium and lipid uptake of macrophages, which transform into foam cells and contribute to atherosclerosis (1214). Furthermore, an increased level of TNF-α, MCP-1, and matrix-metalloproteinase-9 has been described in OSAS patients (8, 11).

Monocytes can be classified into three subsets based on their CD14 and CD16 expression (1517). CD14++CD16 are called “classical” monocytes and comprise the biggest compartment with ∼85% of monocytes. They are professional phagocytes, have the highest capacity to migrate, and secrete the highest amounts of proinflammatory cytokines. CD14+CD16+ “intermediate” monocytes are with 5–10% of the major APCs. The remaining 5–10% constitute CD14dim+CD16+ “nonclassical” monocytes, which are associated with antiviral immunity because of their higher expression of TLR7 and TLR9 (18, 19). It has been hypothesized that both CD16+ subsets emerge from the classical monocyte population and are therefore more differentiated cells (2022). An increase of CD16+ monocytes has been described in many inflammatory conditions as sepsis (23), chronic viral infections (24), rheumatoid arthritis (25), periodontitis (26), and asthma (27). Up to now, nothing is known about the peripheral monocyte composition in OSAS patients.

T cells are crucial players for the release of proinflammatory cytokines. In recent years, it became increasingly evident that T cells are affected by OSAS-induced oxidative stress (2830). Changes of the B, T, and NK cell composition and activation of certain lymphocyte subsets have been described (3133). In detail, a shift to a TH1/TH17 dominant phenotype with a lack of regulatory T cells has been reported in OSAS patients (34, 35). Moreover, an altered phenotype and function of cytotoxic NK cells and CD8+ T cells has been proposed, which is characterized by increased cytotoxicity against endothelial cells (36, 37). As a consequence, vascular damage is done by activated T cells and monocytes, which leads to the development of OSAS-related cardiovascular disease.

The aim of the study was to understand the impact of OSAS on differentiation patterns of circulating monocytes with regard to the three above-described subsets. Furthermore, a detailed analysis of the CD4/CD8 T cell subset composition, especially with respect to the immune checkpoint molecules programmed death 1 (PD-1) and programmed death ligand 1 (PD-L1), was addressed because an upregulated PD-1/PD-L1 cross-talk is known to suppress T cell activation. The study aimed to increase our understanding on immunological changes in these cells and their possible underlying interplay.

All blood donors have signed an informed written consent and were informed about the aims of the study and the use of their samples. Blood samples were collected from healthy donors (n = 20) or OSAS patients (n = 48). Blood was drawn by venipuncture into a sodium citrate containing S-Monovette (Sarstedt, Nümbrecht, Germany). All OSAS patients were treatment naive and analyzed directly after first-time diagnosis. They had a mean age of 50 (±14), a body mass index (BMI) of 29.18 kg/m2 (±5.16), an apnea–hypopnea index (AHI) of 17.8 events/h (±16.96), and a minimal O2 saturation of 88.58% (±4.93). Healthy donors had a mean age of 48 (±16) and a BMI of 24.56 kg/m2 (±3.60).

Within 4 h after blood collection, 20 μl of citrate blood was diluted in 80 μl PBS. Blood cells were stained with following Abs: CD45-PE, CD14-FITC, CD16-BV-510, HLA-DR-allophycocyanin-Cy7, and CD3-PerCP (all from BioLegend, San Diego, CA). After 25-min staining in the dark, 650 μl RBC Lysis Buffer (BioLegend) was added to the samples and incubated for another 20 min. Subsequently, suspension was centrifuged at 400 × g for 5 min, and supernatant was discarded. Cell pellet was resuspended in 100 μl fresh PBS and ready for FACS analysis.

PBMC were isolated from the remaining blood by density gradient centrifugation in Biocoll (Biochrom, Berlin, Germany) at 400 × g for 20 min. The upper PBS/plasma layer was removed and discarded. The PBMC layer was carefully harvested and transferred to a new 15-ml tube and washed once with PBS. The supernatant was discarded again and the PBMC pellet was resuspended in 1 ml PBS. For analysis of T cell subsets, 100 μl of the cell suspension was incubated with two different Ab mixtures: 1) CD3-PerCP, CD4-PE-Cy7, CD8-BV-510, PD-1-PE, PD-L1-allophycocyanin, CD45RA-allophycocyanin-Cy7, and CCR7-BV421 and 2) CD3-PerCP, CD4-PE-Cy7, CD8-BV-510, PD-L1-allophycocyanin, CCR6-allophycocyanin-Cy7, CXCR3-BV421, CD39-PE, and CD25-FITC. After 25-min staining in the dark, cells were washed with PBS and centrifuged at 400 × g for 5 min. Cell pellets were resuspended in 200 μl PBS and prepared for FACS analysis.

Flow cytometry was performed with a MACSQuant 10 flow cytometer (Miltenyi Biotec, Bergisch-Gladbach, Germany), and data were analyzed using the FlowJo software version 10.0 (FlowJo, Ashland, OR). Subsequent to Ab titrations and compensations, at least 100,000 CD45+ leukocytes were analyzed for whole blood measurements. Gating of monocyte subsets was performed as described by Marimuthu and colleagues (38) and shown in Fig. 1. In brief, CD45 was used as a pan leukocyte marker to facilitate whole blood measurement and exclusion of debris. After doublet exclusion, monocytes were first roughly gated by their forward scatter (FSC)/side scatter (SSC) characteristics and the positivity for CD14 and CD16. Neutrophil granulocytes, NK cells, and B cells were excluded with the help of HLA-DR and CD14, respectively. The remaining real monocytes were then subgated into CD14++CD16 (classical), CD14++CD16+ (intermediate), and CD14dim+CD16+ (nonclassical) monocytes.

For T cell analysis, 100,000 events within the PBMC gate were measured. Gating scheme of T cell subpopulations is shown in Fig. 2.

Statistical analyses were performed with GraphPad Prism Version 7.0f (GraphPad Software, San Diego, CA). The mean and SEM are presented. The differences between groups were determined after testing for Gaussian distribution (normality tests) and applying parametric (Student t test) or nonparametric one-way ANOVA with Bonferroni post hoc test. The correlation between parameters was calculated using multivariate regression with the Pearson correlation coefficient: *p < 0.05, **p < 0.01, and ***p < 0.001. Additional statistical details are given in the respective figure legends, when appropriate.

Variations in replicate numbers for certain parameters were caused by too old blood samples for proper monocyte analysis or too low PBMC numbers for reputable T cell subset characterization, respectively.

Our investigations identified strong alterations of the abundance of all three monocyte subsets in OSAS patients compared with healthy donors (Fig. 1). Classical monocytes were significantly decreased, whereas intermediate and nonclassical subsets were significantly increased (Fig. 2). Furthermore, based on the severity of monocyte alteration, OSAS patients could be subdivided into three distinct groups. Seven patients showed quite normal percentages of the monocyte subsets and no or only mild alterations in comparison with healthy donors (Fig. 3A). Most patients (n = 29) revealed a moderate decrease of classical monocytes accompanied by an increase of intermediate monocytes. Nine patients showed a severe drop of classical monocytes with a strong increase of intermediate and nonclassical monocytes (Fig. 3A, Table I). There was a positive correlation between the severity of the monocyte alterations and the BMI of the patients (p = 0.039; Fig. 4). In contrast, AHI, minimal O2 saturation, or age was not found to be correlated with percentages of monocyte subsets. Therefore, all clinical parameters were not significantly different between the defined subgroups (Table I).

FIGURE 1.

Gating strategy of the flow cytometric analysis of monocyte subsets. Leukocytes were measured and gated by expression of CD45. After doublet exclusion, monocytes were first roughly gated by their forward scatter (FSC)/side scatter (SSC) characteristics and further on by their CD14 and CD16 positivity. NK cells and neutrophil granulocytes were excluded by their missing HLA-DR expression. Remaining B cells were excluded by the help of their lack of CD14 expression. Monocyte subsets were finally subdivided into CD14++CD16 (classical), CD14++CD16+ (intermediate), and CD14dim+CD16+ (nonclassical) monocytes.

FIGURE 1.

Gating strategy of the flow cytometric analysis of monocyte subsets. Leukocytes were measured and gated by expression of CD45. After doublet exclusion, monocytes were first roughly gated by their forward scatter (FSC)/side scatter (SSC) characteristics and further on by their CD14 and CD16 positivity. NK cells and neutrophil granulocytes were excluded by their missing HLA-DR expression. Remaining B cells were excluded by the help of their lack of CD14 expression. Monocyte subsets were finally subdivided into CD14++CD16 (classical), CD14++CD16+ (intermediate), and CD14dim+CD16+ (nonclassical) monocytes.

Close modal
FIGURE 2.

Gating strategy of the flow cytometric analysis of peripheral T lymphocyte subsets. PBMC were first roughly gated by FSC/SSC characteristics, and doublets were excluded. The CD3+ population represents T cells. Another CD4dim+CD3 population represents monocytes. CD3+ T cells were further subgated into CD4 or CD8 positives. T cell differentiation state was classified by the help of CD45RA and CCR7 into naive (N), effector (E), effector memory (EM), and central memory for CD4+ and CD8+. CD4+ Th cell subsets were further subdivided by CXCR3 and CCR6. Percentages of PD-1+ and PD-L1+ as well as the expression intensities (MFI) of both markers were determined for CD4+ and CD8+ T cells. PD-L1 expression intensity was also determined for monocytes.

FIGURE 2.

Gating strategy of the flow cytometric analysis of peripheral T lymphocyte subsets. PBMC were first roughly gated by FSC/SSC characteristics, and doublets were excluded. The CD3+ population represents T cells. Another CD4dim+CD3 population represents monocytes. CD3+ T cells were further subgated into CD4 or CD8 positives. T cell differentiation state was classified by the help of CD45RA and CCR7 into naive (N), effector (E), effector memory (EM), and central memory for CD4+ and CD8+. CD4+ Th cell subsets were further subdivided by CXCR3 and CCR6. Percentages of PD-1+ and PD-L1+ as well as the expression intensities (MFI) of both markers were determined for CD4+ and CD8+ T cells. PD-L1 expression intensity was also determined for monocytes.

Close modal
FIGURE 3.

Flow cytometric analysis of monocytes. (A) Whole blood analysis revealed a decrease of classical monocytes (CM) and an increase of CD16+ intermediate monocytes (IM) and nonclassical monocytes (NCM) in OSAS patients (n = 45) compared with healthy donors (n = 10). According to the severity of monocyte alteration, patients could be divided into three groups. (B) FACS analysis of PBMC from OSAS patients (n = 37) and healthy donors (n = 14) revealed an increased PD-L1 expression on monocytes from OSAS patients, which was correlated with the severity of monocyte alteration by tendency. (C) Complex formation of monocytes and T cells analyzed by flow cytometry in whole blood. Percentage of monocyte-complexed T cells was determined by CD14+ cells within the CD3+ T cell subpopulation (n = 8 healthy donors; 24 OSAS). *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001.

FIGURE 3.

Flow cytometric analysis of monocytes. (A) Whole blood analysis revealed a decrease of classical monocytes (CM) and an increase of CD16+ intermediate monocytes (IM) and nonclassical monocytes (NCM) in OSAS patients (n = 45) compared with healthy donors (n = 10). According to the severity of monocyte alteration, patients could be divided into three groups. (B) FACS analysis of PBMC from OSAS patients (n = 37) and healthy donors (n = 14) revealed an increased PD-L1 expression on monocytes from OSAS patients, which was correlated with the severity of monocyte alteration by tendency. (C) Complex formation of monocytes and T cells analyzed by flow cytometry in whole blood. Percentage of monocyte-complexed T cells was determined by CD14+ cells within the CD3+ T cell subpopulation (n = 8 healthy donors; 24 OSAS). *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001.

Close modal
Table I.
Percentage of each monocyte subset and clinical data of OSAS patients as they were classified into three groups based on the severity of monocyte alterations
Mild AlterationModerate AlterationSevere Alteration
Classical monocytes (%) 86.07 ± 3.52 69.57 ± 8.24 43.79 ± 9.87 
Intermediate monocytes (%) 9.23 ± 2.80 20.62 ± 7.74 35.70 ± 9.46 
Nonclassical monocytes (%) 4.67 ± 2.27 9.59 ± 2.93 20.41 ± 5.28 
AHI (per h) 12.74 ± 6.31 19.02 ± 19.92 19.00 ± 8.83 
Minimal O2 saturation (%) 90.14 ± 3.10 89.5 ± 4.42 85.1 ± 7.11 
BMI (kg/m226.35 ± 5.00 29.50 ± 4.08 31.25 ± 4.32 
Age (y) 45 ± 15 51 ± 14 53 ± 13 
Mild AlterationModerate AlterationSevere Alteration
Classical monocytes (%) 86.07 ± 3.52 69.57 ± 8.24 43.79 ± 9.87 
Intermediate monocytes (%) 9.23 ± 2.80 20.62 ± 7.74 35.70 ± 9.46 
Nonclassical monocytes (%) 4.67 ± 2.27 9.59 ± 2.93 20.41 ± 5.28 
AHI (per h) 12.74 ± 6.31 19.02 ± 19.92 19.00 ± 8.83 
Minimal O2 saturation (%) 90.14 ± 3.10 89.5 ± 4.42 85.1 ± 7.11 
BMI (kg/m226.35 ± 5.00 29.50 ± 4.08 31.25 ± 4.32 
Age (y) 45 ± 15 51 ± 14 53 ± 13 

Clinical parameters as median AHI, minimal O2 saturation, BMI, and age of the patients are shown for each defined group. No significant differences were found.

FIGURE 4.

Correlation analysis between the different parameters measured in OSAS patients (n = 48). A multivariate regression with the Pearson correlation coefficient was performed. The correlation coefficient (r) and p values are given for each pair. p < 0.05 was considered significant.

FIGURE 4.

Correlation analysis between the different parameters measured in OSAS patients (n = 48). A multivariate regression with the Pearson correlation coefficient was performed. The correlation coefficient (r) and p values are given for each pair. p < 0.05 was considered significant.

Close modal

The PD-L1 expression of monocytes was also analyzed during FACS analysis of PBMCs. Monocytes were gated by their FSC/SSC characteristics and further on by the help of their low positivity for CD4 and missing CD3 expression (Fig. 2). Our data revealed a significantly higher PD-L1 expression on monocytes of OSAS patients compared with healthy donors (Fig. 3B). The PD-L1 expression level also showed a significant correlation with the severity of the monocyte alterations (p = 0.018). Furthermore, there was a positive correlation between monocytic PDL-1 expression and the AHI (p = 0.02) and the BMI (p = 0.012) of the patients (Fig. 4).

Our analyses also identified increased levels of monocyte/T cell aggregates in the peripheral blood of OSAS patients using flow cytometry. Double staining using anti-CD14 and anti-CD3 Abs represent CD14+CD3+ monocyte/T cell aggregates within the analyzed T cell fraction. The percentage of monocyte-aggregated T cells was significantly increased in OSAS patients compared with healthy donors (Fig. 3C).

Subsequent investigations of T cell subsets in OSAS patients compared with healthy donors were carried out using flow cytometry (Fig. 2). The T cell differentiation from naive to effector, effector memory, and central memory cells was analyzed for CD4+ and CD8+ T cells by specific makers. Moreover, CD4 T helper subsets TH1, TH2, and TH17/22 could be distinguished. The percentages of each subset as well as the PD-1 and PD-L1 expression levels were analyzed.

Our data revealed significantly increased levels of CD4+ effector T cells accompanied by a decrease of effector memory and central memory T cells (Fig. 5A). CD8+ T cells were not significantly altered in OSAS patients (Fig. 5A). Percentages of PD-1+ cells as well as the PD-1 expression intensity (mean fluorescence intensity [MFI]) were significantly increased on CD4+ and CD8+ T cells in OSAS patients, indicating an activated immune phenotype (Fig. 5B). PD-L1 expression was also slightly increased on CD4+ and CD8+ T cells from OSAS patients whereby big individual differences could be observed (Fig. 5C).

FIGURE 5.

Flow cytometric analysis of CD4+ and CD8+ T cell subsets in PBMC from OSAS patients (n = 40) and healthy donors (n = 20). (A) Percentages of naive, effector, effector memory, and central memory T cells within CD4+ or CD8+ T cells. (B) Percentage of PD-1+ and PD-1 expression intensity (MFI) of CD4+ or CD8+ T cells. (C) Percentage of PD-L1+ and PD-L1 expression intensity (MFI) of CD4+ or CD8+ T cells. (D) Percentages of CD4+ Th cell subsets TH1, TH2, or TH17/22. *p ≤ 0.05, ***p ≤ 0.001.

FIGURE 5.

Flow cytometric analysis of CD4+ and CD8+ T cell subsets in PBMC from OSAS patients (n = 40) and healthy donors (n = 20). (A) Percentages of naive, effector, effector memory, and central memory T cells within CD4+ or CD8+ T cells. (B) Percentage of PD-1+ and PD-1 expression intensity (MFI) of CD4+ or CD8+ T cells. (C) Percentage of PD-L1+ and PD-L1 expression intensity (MFI) of CD4+ or CD8+ T cells. (D) Percentages of CD4+ Th cell subsets TH1, TH2, or TH17/22. *p ≤ 0.05, ***p ≤ 0.001.

Close modal

We further questioned whether the grade of monocyte alteration was related to changes we found in T cell subsets. Therefore, patients were subdivided as described earlier into three groups: “mild alteration,” “moderate alteration,” and “severe alteration.” The composition of T cell subsets showed no differences between the groups (data not shown). The PD-1 expression of CD4+ and CD8+ T cells was linked to the grade of monocyte alteration. Only patients with a severe monocyte alteration revealed a significant difference compared with healthy donors in PD-1 expression on CD4+ and CD8+ T cells (Fig. 6). The PD-L1 expression on both T cell subtypes was consequently significantly correlated with the severity of monocyte alterations (p ≤ 0.001). Only OSAS patients with severe monocyte alteration revealed an increased PDL-1 expression on CD4+ T cells (Fig. 6). Of note, CD8+ T cells showed the highest increase of PD-L1 expression in the patient group with severe monocyte alterations, which was also significantly higher than in all other groups.

FIGURE 6.

Flow cytometric analysis of the PD-1 and PD-L1 expression intensities (MFI) on CD4+ and CD8+ T cells from PBMC of healthy donors and OSAS patients classified according to the severity of monocyte alteration. n = 20 healthy donors; 7 mild alteration, 29 moderate alteration, and 9 severe alteration. *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001.

FIGURE 6.

Flow cytometric analysis of the PD-1 and PD-L1 expression intensities (MFI) on CD4+ and CD8+ T cells from PBMC of healthy donors and OSAS patients classified according to the severity of monocyte alteration. n = 20 healthy donors; 7 mild alteration, 29 moderate alteration, and 9 severe alteration. *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001.

Close modal

The present study was undertaken to investigate the impact of OSAS on percentages of monocyte subsets, monocyte–T cell complex formation, and T cell immune alterations, distinguishing between CD4+ and CD8+ T cells. Additionally, the expression levels of PD-1 and PD-L1 on the peripheral T cells of OSAS patients and healthy donors were analyzed for CD3+CD4+ and the CD3+CD8+ subsets and monocytes using flow cytometry.

Under healthy conditions, around 85% of monocytes are classical CD14+CD16- monocytes, which are characterized by phagocytosis, endothelial transmigration, and production of proinflammatory cytokines. The remaining 15% are linked to more specialized Ag presentation functions, defense against viruses, and patrolling behavior (7). Under various inflammatory conditions, an increase of CD16+ intermediate and nonclassical subsets has been reported (15). These subsets are generally termed proinflammatory because of their high production of TNF-α and IL-1β. In our study, we showed for the first time, to our knowledge, a massive decrease of classical monocytes accompanied by an increase of CD16+ subsets in OSAS patients (Fig. 3A). A similar high percentage of intermediate monocytes was up to now only reported in asthma (27). A possible explanation is OSAS-related oxidative stress, followed by upregulation of proinflammatory transcription factors namely NF-κB and HIF-1α (39).

Another important factor is obesity. We found a positive correlation between the severity of monocyte alterations with the BMI of the patients. The World Health Organization reported that half of the European population is overweight, and ∼30% of those are obese. In our patient cohort, 85% were overweight, and 40% of these were obese. Of note, an increase of the intermediate and nonclassical monocyte subsets has already been shown in obesity (4042). A coherence of obesity, OSAS, and atherosclerosis has been well described in recent years (7, 39, 43, 44). Our data corroborate this thesis as we found a positive correlation between BMI and AHI (p = 0.041) and a negative correlation between BMI and O2 saturation by tendency (p = 0.076; Fig. 4). OSAS evokes less physical activity and metabolic dysfunction, which promotes obesity. Obesity, in contrast, is a major risk factor for OSAS (44). Both conditions promote a higher presence of proinflammatory mediators and are therefore classified as low-grade systemic inflammation. Because of mutual molecular signaling pathways, OSAS and obesity are likely to act synergistically as factors of immune disturbance. An additional therapy for body weight reduction might therefore be helpful for OSAS patients.

Furthermore, we found an increase of PD-L1 expression on monocytes (Fig. 3B), which has been described before (45). The monocytic PD-L1 expression level was the only parameter correlating with the AHI in our patient cohort (p = 0.02; Fig. 4). It was additionally correlated with the severity of monocyte alteration. The monocytic PD-L1 expression also showed a positive correlation with the PD-L1 expression on CD4+ and CD8+ T cells, which was likewise highest in patients with severe monocyte alterations (Fig. 6). Intermittent hypoxia has been linked to an upregulation of HIF-1α, which induces an increase of PD-L1 on monocytes (30) and T cells (29).

Our study revealed significantly increased T cell–monocyte aggregates in OSAS patients compared with healthy donors (Fig. 3C). A recent study proposed that these aggregates are a crucial marker for any type of immune perturbations (46). These findings could also be a hint for increased communication between these cells, causing an altered PD-1/PD-L1–mediated immune regulation. We found a significant increase of effector CD4+ T cells and a higher expression of PD-1 on CD4+ and CD8+ T cells (Fig. 5B). PD-1 expression is a natural consequence of T cell activation. Engagement of PD-1 on T cell surface inhibits T cell proliferation and effector functions (47, 48). PD-L1 on monocytes, in contrast, promotes a crucial mechanism for the regulation of T cell responses preventing overreaction. PD-L1+ T cells have also been hypothesized to perform regulatory functions by themselves (49, 50). An imbalanced activation of the PD-1/PD-L1 pathway can, however, ultimately lead to the development of chronic inflammations and related diseases. In fact, an imbalanced PD-L1 signaling has been associated with chronic viral or bacterial infections (51, 52). Impaired T cell functions caused by hypoxia-induced PD-1/PD-L1 cross-talk have been described in OSAS patients (30, 45). The authors explained thereby the higher aggressiveness of cancer in OSAS patients, and PD-L1–mediated immune suppression could also be a protective mechanism to avoid substantial damage to the vascular system and related diseases.

Several studies reported an increase of TH1/TH17 cells accompanied by a decrease of regulatory T cells, which leads to a shift to an overall proinflammatory milieu (3335). Our data revealed no increase in TH1/TH17 cells. Our findings might be explained by the overall moderate grade of OSAS in our patient cohort as alterations were often linked to the AHI. Changes in these T cell compositions might predominantly occur in patients with severe OSAS.

In conclusion, the current study indicates that OSAS and related obesity induces alterations in the composition of peripheral monocyte subsets and increased PD-L1 expression on monocytes. Furthermore, monocyte alterations were found to be connected to increased T cell activation accompanied by an imbalanced PD-1/PD-L1 cross-talk. However, attention should be paid to the fact that our study cohort was composed mostly of patients with mild to moderate OSAS and that severe monocyte alterations were only found in 9 out of 48 patients. It is possible that other comorbidities are not negligible factors that provoke monocyte subset alterations. Nevertheless, the findings presented in this study provide new insights concerning monocyte-driven immune alterations explaining the higher incidence of infections, cardiovascular diseases, and cancer in OSAS patients.

We are grateful to all members of the Department of Otorhinolaryngology for stimulating and helpful discussions.

This work was supported by the Rudolf-Bartling-Stiftung.

Abbreviations used in this article:

AHI

apnea–hypopnea index

BMI

body mass index

FSC

forward scatter

MFI

mean fluorescence intensity

OSAS

obstructive sleep apnea syndrome

PD-1

programmed death 1

PD-L1

programmed death ligand 1

SSC

side scatter.

1
Rasch
,
B.
,
J.
Born
.
2013
.
About sleep’s role in memory.
Physiol. Rev.
93
:
681
766
.
2
Myung
,
J.
,
D.
Wu
,
V.
Simonneaux
,
T. J.
Lane
.
2018
.
Strong circadian rhythms in the choroid plexus: implications for sleep-independent brain metabolite clearance.
J. Exp. Neurosci.
12
: 1179069518783762.
3
Xie
,
H.
,
J.
Yin
,
Y.
Bai
,
H.
Peng
,
X.
Zhou
,
J.
Bai
.
2019
.
Differential expression of immune markers in the patients with obstructive sleep apnea/hypopnea syndrome.
Eur. Arch. Otorhinolaryngol.
276
:
735
744
.
4
Muehlroth
,
B. E.
,
B.
Rasch
,
M.
Werkle-Bergner
.
2020
.
Episodic memory consolidation during sleep in healthy aging.
Sleep Med. Rev.
52
: 101304.
5
Kapur
,
V. K.
,
D. H.
Auckley
,
S.
Chowdhuri
,
D. C.
Kuhlmann
,
R.
Mehra
,
K.
Ramar
,
C. G.
Harrod
.
2017
.
Clinical practice guideline for diagnostic testing for adult obstructive sleep apnea: an American academy of sleep medicine clinical practice guideline.
J. Clin. Sleep Med.
13
:
479
504
.
6
Ryan
,
S.
,
C. T.
Taylor
,
W. T.
McNicholas
.
2009
.
Systemic inflammation: a key factor in the pathogenesis of cardiovascular complications in obstructive sleep apnoea syndrome?
Postgrad. Med. J.
85
:
693
698
.
7
Kapellos
,
T. S.
,
L.
Bonaguro
,
I.
Gemünd
,
N.
Reusch
,
A.
Saglam
,
E. R.
Hinkley
,
J. L.
Schultze
.
2019
.
Human monocyte subsets and phenotypes in major chronic inflammatory diseases.
Front. Immunol.
10
:
2035
.
8
Minoguchi
,
K.
,
T.
Tazaki
,
T.
Yokoe
,
H.
Minoguchi
,
Y.
Watanabe
,
M.
Yamamoto
,
M.
Adachi
.
2004
.
Elevated production of tumor necrosis factor-alpha by monocytes in patients with obstructive sleep apnea syndrome.
Chest
126
:
1473
1479
.
9
Dyugovskaya
,
L.
,
P.
Lavie
,
L.
Lavie
.
2003
.
Phenotypic and functional characterization of blood gammadelta T cells in sleep apnea.
Am. J. Respir. Crit. Care Med.
168
:
242
249
.
10
Arnaud
,
C.
,
M.
Dematteis
,
J. L.
Pepin
,
J. P.
Baguet
,
P.
Lévy
.
2009
.
Obstructive sleep apnea, immuno-inflammation, and atherosclerosis.
Semin. Immunopathol.
31
:
113
125
.
11
Tamaki
,
S.
,
M.
Yamauchi
,
A.
Fukuoka
,
K.
Makinodan
,
N.
Koyama
,
K.
Tomoda
,
M.
Yoshikawa
,
H.
Kimura
.
2009
.
Nocturnal hypoxic stress activates invasive ability of monocytes in patients with obstructive sleep apnoea syndrome.
Respirology
14
:
689
694
.
12
Zhou
,
J.
,
W.
Bai
,
Q.
Liu
,
J.
Cui
,
W.
Zhang
.
2018
.
Intermittent hypoxia enhances THP-1 monocyte adhesion and chemotaxis and promotes M1 macrophage polarization via RAGE.
BioMed Res. Int.
2018
: 1650456.
13
Tamaki
,
S.
,
M.
Yamauchi
,
A.
Fukuoka
,
K.
Makinodan
,
N.
Koyama
,
K.
Tomoda
,
M.
Yoshikawa
,
H.
Kimura
.
2009
.
Production of inflammatory mediators by monocytes in patients with obstructive sleep apnea syndrome.
Intern. Med.
48
:
1255
1262
.
14
Inonu Koseoglu
,
H.
,
A. C.
Pazarli
,
A.
Kanbay
,
O.
Demir
.
2018
.
Monocyte count/HDL cholesterol ratio and cardiovascular disease in patients with obstructive sleep apnea syndrome: a multicenter study.
Clin. Appl. Thromb. Hemost.
24
:
139
144
.
15
Wong
,
K. L.
,
W. H.
Yeap
,
J. J.
Tai
,
S. M.
Ong
,
T. M.
Dang
,
S. C.
Wong
.
2012
.
The three human monocyte subsets: implications for health and disease.
Immunol. Res.
53
:
41
57
.
16
Patel
,
A. A.
,
Y.
Zhang
,
J. N.
Fullerton
,
L.
Boelen
,
A.
Rongvaux
,
A. A.
Maini
,
V.
Bigley
,
R. A.
Flavell
,
D. W.
Gilroy
,
B.
Asquith
, et al
.
2017
.
The fate and lifespan of human monocyte subsets in steady state and systemic inflammation.
J. Exp. Med.
214
:
1913
1923
.
17
Ziegler-Heitbrock
,
L.
2015
.
Blood monocytes and their subsets: established features and open questions.
Front. Immunol.
6
:
423
.
18
Boyette
,
L. B.
,
C.
Macedo
,
K.
Hadi
,
B. D.
Elinoff
,
J. T.
Walters
,
B.
Ramaswami
,
G.
Chalasani
,
J. M.
Taboas
,
F. G.
Lakkis
,
D. M.
Metes
.
2017
.
Phenotype, function, and differentiation potential of human monocyte subsets.
PLoS One
12
: e0176460.
19
Cros
,
J.
,
N.
Cagnard
,
K.
Woollard
,
N.
Patey
,
S. Y.
Zhang
,
B.
Senechal
,
A.
Puel
,
S. K.
Biswas
,
D.
Moshous
,
C.
Picard
, et al
.
2010
.
Human CD14dim monocytes patrol and sense nucleic acids and viruses via TLR7 and TLR8 receptors.
Immunity
33
:
375
386
.
20
Ancuta
,
P.
,
K. Y.
Liu
,
V.
Misra
,
V. S.
Wacleche
,
A.
Gosselin
,
X.
Zhou
,
D.
Gabuzda
.
2009
.
Transcriptional profiling reveals developmental relationship and distinct biological functions of CD16+ and CD16- monocyte subsets.
BMC Genomics
10
:
403
.
21
Jakubzick
,
C. V.
,
G. J.
Randolph
,
P. M.
Henson
.
2017
.
Monocyte differentiation and antigen-presenting functions.
Nat. Rev. Immunol.
17
:
349
362
.
22
Coillard
,
A.
,
E.
Segura
.
2019
.
In vivo differentiation of human monocytes.
Front. Immunol.
10
:
1907
.
23
Skrzeczyñska
,
J.
,
K.
Kobylarz
,
Z.
Hartwich
,
M.
Zembala
,
J.
Pryjma
.
2002
.
CD14+CD16+ monocytes in the course of sepsis in neonates and small children: monitoring and functional studies.
Scand. J. Immunol.
55
:
629
638
.
24
Azeredo
,
E. L.
,
P. C.
Neves-Souza
,
A. R.
Alvarenga
,
S. R.
Reis
,
A.
Torrentes-Carvalho
,
S. M.
Zagne
,
R. M.
Nogueira
,
L. M.
Oliveira-Pinto
,
C. F.
Kubelka
.
2010
.
Differential regulation of toll-like receptor-2, toll-like receptor-4, CD16 and human leucocyte antigen-DR on peripheral blood monocytes during mild and severe dengue fever.
Immunology
130
:
202
216
.
25
Rossol
,
M.
,
S.
Kraus
,
M.
Pierer
,
C.
Baerwald
,
U.
Wagner
.
2012
.
The CD14(bright) CD16+ monocyte subset is expanded in rheumatoid arthritis and promotes expansion of the Th17 cell population.
Arthritis Rheum.
64
:
671
677
.
26
Nagasawa
,
T.
,
H.
Kobayashi
,
M.
Aramaki
,
M.
Kiji
,
S.
Oda
,
Y.
Izumi
.
2004
.
Expression of CD14, CD16 and CD45RA on monocytes from periodontitis patients.
J. Periodontal Res.
39
:
72
78
.
27
Moniuszko
,
M.
,
A.
Bodzenta-Lukaszyk
,
K.
Kowal
,
D.
Lenczewska
,
M.
Dabrowska
.
2009
.
Enhanced frequencies of CD14++CD16+, but not CD14+CD16+, peripheral blood monocytes in severe asthmatic patients.
Clin. Immunol.
130
:
338
346
.
28
Lavie
,
L.
,
L.
Dyugovskaya
,
A.
Polyakov
.
2008
.
Biology of peripheral blood cells in obstructive sleep apnea--the tip of the iceberg.
Arch. Physiol. Biochem.
114
:
244
254
.
29
Kurobe
,
H.
,
M.
Urata
,
M.
Ueno
,
M.
Ueki
,
S.
Ono
,
Y.
Izawa-Ishizawa
,
Y.
Fukuhara
,
Y.
Lei
,
A. M.
Ripen
,
T.
Kanbara
, et al
.
2010
.
Role of hypoxia-inducible factor 1alpha in T cells as a negative regulator in development of vascular remodeling.
Arterioscler. Thromb. Vasc. Biol.
30
:
210
217
.
30
Cubillos-Zapata
,
C.
,
J.
Avendaño-Ortiz
,
E.
Hernandez-Jimenez
,
V.
Toledano
,
J.
Casas-Martin
,
A.
Varela-Serrano
,
M.
Torres
,
I.
Almendros
,
R.
Casitas
,
I.
Fernández-Navarro
, et al
.
2017
.
Hypoxia-induced PD-L1/PD-1 crosstalk impairs T-cell function in sleep apnoea.
Eur. Respir. J.
50
: 1700833.
31
Domagała-Kulawik
,
J.
,
I.
Osińska
,
A.
Piechuta
,
P.
Bielicki
,
T.
Skirecki
.
2015
.
T, B, and NKT cells in systemic inflammation in obstructive sleep apnoea.
Mediators Inflamm.
2015
: 161579.
32
Domagała-Kulawik
,
J.
,
I.
Kwiecień
,
P.
Bielicki
,
T.
Skirecki
.
2019
.
Fas-positive lymphocytes are associated with systemic inflammation in obstructive sleep apnea syndrome.
Sleep Breath.
23
:
673
678
.
33
Ye
,
J.
,
H.
Liu
,
G.
Zhang
,
P.
Li
,
Z.
Wang
,
S.
Huang
,
Q.
Yang
,
Y.
Li
.
2012
.
The treg/th17 imbalance in patients with obstructive sleep apnoea syndrome.
Mediators Inflamm.
2012
: 815308.
34
Tan
,
H. L.
,
D.
Gozal
,
Y.
Wang
,
H. P.
Bandla
,
R.
Bhattacharjee
,
R.
Kulkarni
,
L.
Kheirandish-Gozal
.
2013
.
Alterations in circulating T-cell lymphocyte populations in children with obstructive sleep apnea.
Sleep (Basel)
36
:
913
922
.
35
Anderson
,
M. E.
 Jr.
,
Z. S.
Buchwald
,
J.
Ko
,
R.
Aurora
,
T.
Sanford
.
2014
.
Patients with pediatric obstructive sleep apnea show altered T-cell populations with a dominant TH17 profile.
Otolaryngol. Head Neck Surg.
150
:
880
886
.
36
Hernández-Jiménez
,
E.
,
C.
Cubillos-Zapata
,
V.
Toledano
,
R.
Pérez de Diego
,
I.
Fernández-Navarro
,
R.
Casitas
,
C.
Carpio
,
J.
Casas-Martín
,
J.
Valentín
,
A.
Varela-Serrano
, et al
.
2017
.
Monocytes inhibit NK activity via TGF-β in patients with obstructive sleep apnoea.
Eur. Respir. J.
49
:
1602456
.
37
Dyugovskaya
,
L.
,
P.
Lavie
,
M.
Hirsh
,
L.
Lavie
.
2005
.
Activated CD8+ T-lymphocytes in obstructive sleep apnoea.
Eur. Respir. J.
25
:
820
828
.
38
Marimuthu
,
R.
,
H.
Francis
,
S.
Dervish
,
S. C. H.
Li
,
H.
Medbury
,
H.
Williams
.
2018
.
Characterization of human monocyte subsets by whole blood flow cytometry analysis.
J. Vis. Exp.
140
:
579471
. DOI: 10.3791/57941.
39
Ryan
,
S.
,
C.
Arnaud
,
S. F.
Fitzpatrick
,
J.
Gaucher
,
R.
Tamisier
,
J.-L.
Pépin
.
2019
.
Adipose tissue as a key player in obstructive sleep apnoea.
Eur. Respir. Rev.
28
: 190006.
40
Poitou
,
C.
,
E.
Dalmas
,
M.
Renovato
,
V.
Benhamo
,
F.
Hajduch
,
M.
Abdennour
,
J. F.
Kahn
,
N.
Veyrie
,
S.
Rizkalla
,
W. H.
Fridman
, et al
.
2011
.
CD14dimCD16+ and CD14+CD16+ monocytes in obesity and during weight loss: relationships with fat mass and subclinical atherosclerosis.
Arterioscler. Thromb. Vasc. Biol.
31
:
2322
2330
.
41
Devêvre
,
E. F.
,
M.
Renovato-Martins
,
K.
Clément
,
C.
Sautès-Fridman
,
I.
Cremer
,
C.
Poitou
.
2015
.
Profiling of the three circulating monocyte subpopulations in human obesity.
J. Immunol.
194
:
3917
3923
.
42
Russo
,
L.
,
C. N.
Lumeng
.
2018
.
Properties and functions of adipose tissue macrophages in obesity.
Immunology
155
:
407
417
.
43
Gaines
,
J.
,
A. N.
Vgontzas
,
J.
Fernandez-Mendoza
,
S. L.
Calhoun
,
F.
He
,
D.
Liao
,
M. D.
Sawyer
,
E. O.
Bixler
.
2016
.
Inflammation mediates the association between visceral adiposity and obstructive sleep apnea in adolescents.
Am. J. Physiol. Endocrinol. Metab.
311
:
E851
E858
.
44
Almendros
,
I.
,
M. A.
Martinez-Garcia
,
R.
Farré
,
D.
Gozal
.
2020
.
Obesity, sleep apnea, and cancer.
Int. J. Obes.
44
:
1653
1667
.
45
Cubillos-Zapata
,
C.
,
I.
Almendros
,
E.
Díaz-García
,
V.
Toledano
,
R.
Casitas
,
R.
Galera
,
E.
López-Collazo
,
R.
Farre
,
D.
Gozal
,
F.
García-Rio
.
2020
.
Differential effect of intermittent hypoxia and sleep fragmentation on PD-1/PD-L1 upregulation.
Sleep
43
: zsz285.
46
Burel
,
J. G.
,
M.
Pomaznoy
,
C. S.
Lindestam Arlehamn
,
D.
Weiskopf
,
R.
da Silva Antunes
,
Y.
Jung
,
M.
Babor
,
V.
Schulten
,
G.
Seumois
,
J. A.
Greenbaum
, et al
.
2019
.
Circulating T cell-monocyte complexes are markers of immune perturbations.
eLife
8
: e46045.
47
Francisco
,
L. M.
,
P. T.
Sage
,
A. H.
Sharpe
.
2010
.
The PD-1 pathway in tolerance and autoimmunity.
Immunol. Rev.
236
:
219
242
.
48
Heinzel
,
S.
,
J. M.
Marchingo
,
M. B.
Horton
,
P. D.
Hodgkin
.
2018
.
The regulation of lymphocyte activation and proliferation.
Curr. Opin. Immunol.
51
:
32
38
.
49
Wu
,
S.-P.
,
R.-Q.
Liao
,
H.-Y.
Tu
,
W.-J.
Wang
,
Z.-Y.
Dong
,
S.-M.
Huang
,
W.-B.
Guo
,
L.-Y.
Gou
,
H.-W.
Sun
,
Q.
Zhang
, et al
.
2018
.
Stromal PD-L1-positive regulatory T cells and PD-1-positive CD8-positive T cells define the response of different subsets of non-small cell lung cancer to PD-1/PD-L1 blockade immunotherapy.
J. Thorac. Oncol.
13
:
521
532
.
50
Liu
,
Y.
,
R.
Carlsson
,
M.
Comabella
,
J.
Wang
,
M.
Kosicki
,
B.
Carrion
,
M.
Hasan
,
X.
Wu
,
X.
Montalban
,
M. H.
Dziegiel
, et al
.
2014
.
FoxA1 directs the lineage and immunosuppressive properties of a novel regulatory T cell population in EAE and MS.
Nat. Med.
20
:
272
282
.
51
McKendry
,
R. T.
,
C. M.
Spalluto
,
H.
Burke
,
B.
Nicholas
,
D.
Cellura
,
A.
Al-Shamkhani
,
K. J.
Staples
,
T. M.
Wilkinson
.
2016
.
Dysregulation of antiviral function of CD8(+) T cells in the chronic obstructive pulmonary disease lung. Role of the PD-1-PD-L1 axis.
Am. J. Respir. Crit. Care Med.
193
:
642
651
.
52
Rutigliano
,
J. A.
,
S.
Sharma
,
M. Y.
Morris
,
T. H.
Oguin
III
,
J. L.
McClaren
,
P. C.
Doherty
,
P. G.
Thomas
.
2014
.
Highly pathological influenza A virus infection is associated with augmented expression of PD-1 by functionally compromised virus-specific CD8+ T cells.
J. Virol.
88
:
1636
1651
.

The authors have no financial conflicts of interest.