• No results found

University of Groningen Adaptation and Modulation of Memory and Regulatory T Cells in Pregnancy Kieffer, Tom Eduard Christiaan

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Adaptation and Modulation of Memory and Regulatory T Cells in Pregnancy Kieffer, Tom Eduard Christiaan"

Copied!
39
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

Adaptation and Modulation of Memory and Regulatory T Cells in Pregnancy

Kieffer, Tom Eduard Christiaan

DOI:

10.33612/diss.97355536

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date: 2019

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Kieffer, T. E. C. (2019). Adaptation and Modulation of Memory and Regulatory T Cells in Pregnancy. Rijksuniversiteit Groningen. https://doi.org/10.33612/diss.97355536

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

7

Decidual Memory T Cell Subsets and

Memory T Cell Stimulatory Cytokines

in Early- and Late-Onset Preeclampsia

Submitted

Tom E.C. Kieffer

1

Anne Laskewitz

2

Annegé Vledder

1

Sicco A. Scherjon

1

Marijke M. Faas

2

Jelmer R. Prins

1

1Department of Obstetrics and Gynaecology, University Medical Center Groningen,

University of Groningen, Groningen, the Netherlands

2Division of Medical Biology, Department of Pathology and Medical Biology,

University Medical Center Groningen, University of Groningen, Groningen, the Netherlands

(3)

ABSTRACT

Preeclampsia is a major cause of fetal and maternal mortality and morbidity. The aetiology of preeclampsia is unknown, but disturbed fetal-maternal tolerance, and therewith memory T cells, might be involved. This study gives insight into memory T cell populations and its associated cytokines in the decidual layers in early-onset preeclampsia (EO-PE) and late-onset preeclampsia (LO-PE). Flow cytometry analysis showed lower CD4+ central-memory (CM) cell proportions in the decidua parietalis in LO-PE (p < 0.0001) and EO-PE (p < 0.01) compared to healthy pregnancies. CD8+ memory (p < 0.05) and CD8+ CM (p < 0.01) cell proportions were also lower in the decidua parietalis in EO-PE compared to healthy pregnancies. This was accompanied by higher IL15 (p < 0.05) and IL23 (p < 0.05) mRNA expression and lower IL7 (p < 0.05) mRNA expression in decidua basalis biopsies from EO-PE compared to healthy pregnancies, analysed by qPCR. In addition, higher activated proportions of CD4+ and CD8+ memory cells were found in EO-PE compared to LO-PE (p < 0.05), revealing an altered memory T cell homeostasis in the preeclampsia subtypes. In conclusion, decidual memory T cell proportions, activation, and associated cytokines are altered in preeclampsia, and might be implicated in its pathophysiology.

INTRODUCTION

Increasing evidence shows that memory T lymphocytes (T cells) may be implicated in the pathophysiology of pregnancy complications as preeclampsia1–4. Preeclampsia affects 5-8% of pregnancies and is a major cause of fetal and maternal mortality and morbidity5,6. The complex multifactorial aetiology of preeclampsia is unknown but preeclampsia is characterized by a poorly established or poorly perfused placenta, endothelial cell activation and a disturbed immune balance leading to hyperten-sion and multi organ failure7. Early-onset preeclampsia (EO-PE) and late-onset pree-clampsia (LO-PE) are distinguished by onset before or after 34 weeks of gestation8. For research purposes this distinction is often used, since the pathophysiology and immunopathology, differs between the two subtypes9,10. EO-PE is associated with disturbed placentation and fetal growth restriction, whereas LO-PE seems associated with ageing of the placenta causing reduced intervillous perfusion9,10.

During pregnancy the maternal immune system has to adapt, as immune tolerance is required towards the semi-allogeneic fetus11–13. Insufficient immune tolerance is associated with pregnancy complications as preeclampsia11,12, which is associated with a pro-inflammatory state of the maternal immune system, skewing away from

(4)

immune tolerance towards the more pregnancy threatening T-helper-1 immunity14. Immune tolerance towards fetal antigens should especially be optimal at the fetal-ma-ternal interface where mafetal-ma-ternal immune cells are in direct contact with fetal cells. The decidua is the maternal tissue that interacts with fetal cells, being the uterine lining in which the fetal trophoblasts anchor15. Two distinct decidua subtypes are identified, characterized by a different immunologic response and different distribution of T cell populations16. The decidua parietalis is the maternal tissue around the chorionic sac and is in direct contact with the non-invading chorionic trophoblast15. The decidua basalis is the maternal side of the placenta and is invaded by the extravillous trop-hoblast17. Many different maternal immune cells are present in both decidual layers, including memory T cells3,18–22.

Memory T cells are a subset of T cells that have developed after a previous encoun-ter with an antigen23. They are long-living and provide immediate protection upon re-exposure to the same antigen24. Memory T cell differentiation, survival, and cyto-kine secretion is under the influence of cytocyto-kines such as interleukin-7 (IL7), IL15 and IL2325–27. Different memory T cell populations can be distinguished based on migra-tion pattern, protein expression profile and cytokine secremigra-tion abilities24,28,29. In both the CD4+ and CD8+ cell lineages, central-memory (CM) and effector-memory (EM) cells are identified23,30–32. CM cells circulate in the blood and migrate preferentially to secondary lymphoid organs32. Upon activation, CM cells can proliferate into EM cells, which circulate throughout the body and generate an aggressive response upon a secondary encounter with the memorized antigen23,30. Within the CD8+ memory T cell lineage, tissue resident memory (TRM) cells can be distinguished29,33. TRM cells do not circulate, reside in tissue, and form the first line of defence on a secondary encounter with the antigen29. In the CD4+ cell lineage, TRM cells are less well defined, but detectable at very low levels33. Finally, there is a population of T regulatory (Treg) memory cells, identified by Foxp3 expression and executing immune regulatory and immunosuppressive function28. CD4+ Treg memory cells have been found to be recrui-ted to the reproductive tract and are important for pregnancy success in mice1,34,35, in humans their importance has not been proven yet. CD4+ Treg memory cells are found at the fetal-maternal interface and in the decidua4,36, but higher proportions are found in the peripheral blood in pregnancy complications as preeclampsia and gestational diabetes compared to healthy controls4,37,38. Treg cells are mainly found in the CD4+ cell lineage, but may also exist within the CD8+ cell lineage39,40.

In contrast to their normal function, memory cells may have a function in tolerance in pregnancy, i.e. fetal tolerance. In murine models it has been shown that fetal antigen

(5)

specific memory T cells are generated during pregnancy that persist postpartum, and decrease fetal resorption rates in a subsequent pregnancy with the same male1,41,42. In human decidua, memory T cells, mostly CD8+ EM cells, were found with a different phenotype and function compared to memory T cells in the peripheral blood43–45. With reduced granzyme B and perforin expression, and high PD-1 expression they appear to be silenced, however, they remain responsive upon stimulation and have high IFN-γ expression43,44. Some studies found that the memory T cell populations in the peripheral blood are affected in preeclamptic pregnancies4,46,47, however decidual memory T cells in preeclampsia are not well studied. Clues for a possible development of immunologic memory to generate appropriate fetal-maternal tolerance, and prevent immune associated pregnancy disorders as preeclampsia are provided by epidemiological studies. It has been shown that preeclampsia is associated with primipaternity48–50. In addition, exposure to paternal antigens via semen before conception is associated with a lower risk of preeclampsia51,52. These findings may imply that the maternal immune system has the ability to memorize paternal and fetal antigens and may be able to facilitate a more appropriate immune response in a subsequent pregnancy with a ‘memorized’ partner.

Studies on memory T cells in preeclampsia are limited and clear insight into memory T cells and the memory T cell cytokine milieu in the different decidual layers is lac-king. The aim of this study is to evaluate differences in memory T cell populations and memory T cell associated cytokines in decidual tissue between uncomplicated pregnancies and pregnancies complicated with preeclampsia.

METHODS

Samples

Placentas and membranes were obtained from 15 healthy women, 8 women diagnosed with late-onset preeclampsia and 6 women diagnosed with early-onset preeclampsia (Table 1). All women had singleton pregnancies, a body mass index <35, no diabetes, and no history of smoking, alcohol or drug use during the current pregnancy. Preeclampsia was defined as gestational hypertension (systolic blood pressure ≥ 140 mmHg and / or diastolic blood pressure > 90 mmHg measured in two different measurements after 20 weeks of gestation) accompanied with either proteinuria (> 300 mg / 24 hours), uteroplacental dysfunction, or other maternal organ dysfunction following the International Society for the Study of Hypertension Guideline53. Diagnosis of preeclampsia before 34 weeks of gestation was considered

(6)

early-onset preeclampsia and diagnosis after 34 weeks was considered late-onset preeclampsia following the classification of the International Society for the Study of Hypertension in Pregnancy8. Placental tissue was used for this study according to the code of conduct for responsible use following the guideline from the Federation of Medical Scientific Associations54. This study, including the consent procedure, was approved by the Medical Ethics Review Board of the University Medical Center Groningen (UMCG) (protocol number METc2018/516). The Medical Ethics Review Board of the UMCG approved that no written informed consent is needed for this study and that the objection procedure against use of medical waste material for research purposes is sufficient. The methods were carried out in accordance with the local guidelines of the UMCG.

Lymphocyte isolation

Placentas and membranes were stored in phosphate buffered saline (PBS) at 4°C directly after delivery until analysis for a maximum of 24 hours55. The amnion was torn off manually after which the decidua parietalis was scraped off the chorion using a cell scraper (Corning incorporated, USA). All blood vessels visible by eye were removed. The villi were cut from the decidua basalis. The volume of tissue collected was noted to evaluate cell yield per ml. The decidua parietalis and decidua basalis were digested in a 1:2 dilution in StemProAccutase (Lifetechnologies, USA) using a GentleMACS Dissociator (Miltenyi, Germany). Lymphocytes were isolated from the digested tissue by centrifugation over a Percoll (Sigma-Aldrich, USA) gradient using three different density layers; 68% Percoll (ρ=1.084 g/ml), 45% Percoll (ρ=1.056 g/ ml) and 40% Percoll (ρ=1.050 g/ml). After centrifugation (30 min, 822 g, 20°C) without deceleration, the lymphocyte enriched layer was isolated between the 68% and 45% Percoll layer. Lymphocytes were washed in PBS and centrifuged (5min, 460 g, 20°C). Cells were counted using a Beckman coulter counter.

Flow cytometry

For viability staining, 106 cells were incubated with 0.001% PercP-Cy5.5 Fixable Viability Stain 620 (BD Biosciences, USA) in PBS for 15 minutes at room tempe-rature in the dark and thereafter washed in PBS. To prevent non-specific binding, cells were incubated in 1% Fc-Block (BD Biosciences, Belgium) and 10% mouse serum (Sanquin, The Netherlands) in PBS at room temperature in the dark for 10 minutes. Cells were incubated with APC-H7 anti-CD4 (SK3, BD Biosciences, Belgium), PE-Cy7 anti-CD8 (RPA-T8, BD Biosciences, Belgium), BV510 anti-CD45RO

(7)

(UCHL-1, BD Biosciences, Belgium), APC anti-CD69 (FN50, BD Biosciences, Belgium), BV421 anti-CD103 (BER-ACT8, BD Biosciences,

Table 1. Patient characteristics

  Healthy (n=15) preeclampsia Late-onset (n=8) Early-onset preeclampsia (n=6)

Maternal age (years) 28 (24-30) 29 (26.25-28.75) 26.0 ±

(25.0-2875) Ethnicity (%)

White 86.67 75 100

Assyrian 13.33 25 0

Gestational age (weeks) 39.29

(39.0-39.86) (37.14-38/79)*38.14 30.65 (29.29-31.32)****

Parity 1 (1-2) 1 (1-2) 1 (1-3)

Body Mass Index (kg / m2) 24.30

(20.35-26.07) (21.65-34.96)26.25 (21.03-26.85)23.85

Gestational age at onset of

preeclampsia (weeks) (35.97-38.21)37.21 (28.4-30.71)†††30.0

Diastolic blood pressure (mmHg) 80.0 (72.0-81.0) 94.0

(86.75-99.63)** (89.0-102.5)***98.50

Systolic blood pressure (mmHg) 120.0

(110.0-122.1) (131.3-155.0)**138.8 (140.0-172.8)***152.5

Duration of hypertension (days) 4.5 (1.0-14.0) 4.5 (3.0-7.5)

Random urine protein:creatinine ratio 0.80 (0.60-0.93) 9.35

(1.84-17.85)†† Corticosteroid therapy for fetal lung

maturation (% of patients) 0 0 100****††††

Mode of delivery (%)

Vaginal 66.67 75.0 0*†

Spontaneous 93.33 37.50 0

Induction on maternal indication 0 62.50 0

Induction on fetal indication 6.67 0 0

(8)

  Healthy (n=15) preeclampsia Late-onset (n=8) Early-onset preeclampsia (n=6) Breech presentation 33.33 0 0 Previous CS 66.67 50.0 0 Fetal indication 0 50.0 16.67 Maternal indication 0 0 83.33

Birth weight (gram) 3410

(3080-3680) (2698-3361)3115 1459)****††††1235

(990-% below 10th percentile birthweight 0 25 66.67**

Fetal sex (% female / male) 40 / 60 12.5 / 87.5 66.7 / 33.3

Time between delivery and start of

placental processing (hours) 6.50 (0-17.50) 10.5 (1.38-17.75) (0.38-19.25)17.50

Amount of cells isolated per ml

decidual tissue (106)

Decidua parietalis 0.17 (0.063-0.21) 0.62 (0.34-0.90) 0.11 (0.052-0.39)

Decidua basalis 0.20 (0.076-0.45) 0.63 (0.23-1.0) 0.12 (0.10-0.24)

Data as median with interquartile range; CS, caesarean section; Blood pressure, highest blood pressure measured; Duration of hypertension, time from first notice of hypertension until birth; statistical analysis by Kolmogorov-Smirnov test, one-way ANOVA and Tukey’s post hoc test, Kruskal-Wallis and Dunnett’s test, or Fisher’s exact test. **p < 0.01, *** p < 0.001, **** p < 0.0001 compared to healthy pregnancies; † p < 0.05, †† p < 0.01, ††† p < 0.001, ††††p < 0.0001 compared to late-onset preeclampsia

Belgium), PE anti-CCR7 (150503, BD Biosciences, Belgium) in PBS at 4°C in the dark for 30 minutes. After surface staining, cells were washed with PBS. To allow for intracellular staining, cells were permeabilized and fixed using Transcription Factor buffer set (BD Biosciences, Belgium) following manufacturerBs instructions. Intracellular staining was performed by incubation with Alexa Fluor 488 anti-Foxp3 (236A/E7, BD Biosciences, Belgium) in PBS at 4°C in the dark for 30 minutes. Cells were washed with wash buffer (BD Biosciences, Belgium) and were analysed with a FACSVerseTM flow cytometer (BD Biosciences, USA) using BD FACS SuiteTM software (BD Biosci-ences, USA). FlowJo V10 software (LLC, USA) was used to analyse flow cytometry data. An overview of the gating strategy is displayed in Supplementary Figure S1. Doublets were excluded using forward and sideward scatterplots (Supplementary Fig. S1A). After doublet exclusion, lymphocytes were gated in a forward sideward scatterplot of the single cells (Figure 1A). Lymphocytes that appeared negative in the

(9)

Fixable Viability Stain dot plot were gated as viable lymphocytes (Figure 1A). Within the viable lymphocyte population, CD4+ and CD8+ cell lineages were distinguished (Figure 1A). Within the CD4+ cell population, the total proportion of CD4+ memory cells (CD45RO+CD4+), CD4+ central-memory (CM) cells (CCR7+CD45RO+CD4+), and CD4+ effector-memory (EM) cells (CCR7-CD45RO+CD4+) were identified (Figure 1B). CD4+ tissue resident memory (TRM) cells (CD103+CD45RO+CD4+) were iden-tified by their expression of CD103 (Figure 1C). Treg CD4+ memory cell propor-tions were determined within the CD4+ memory cell subset by the proportion of Foxp3 expressing CD4+ memory cells (Figure 1D). Foxp3 expression was deter-mined on all the different CD4+ memory cell subsets (Supplementary Figure S1G-1J). The CD69 positive cell population was gated in all different CD4+ memory cell subsets to analyse activated cells (Figure 2A). CD8+ cells were gated following a similar strategy. Within the CD8+ cell population, the total proportion of CD8+ memory cells (CM+EM) (CD45RO+CD8+), CD8+ CM cells (CCR7+CD45RO+CD8+), and CD8+ EM cells (CCR7-CD45RO+CD8+) were identified (Figure 3A). Within the CD8+ cell population, CD8+ TRM cells (CD103+CD45RO+CD8+) were identified by expression of CD103 (Figure 3B). No Foxp3+CD8+ memory cells were observed in the decidual layers. The CD69 positive cell population was gated in all different memory CD8+ cell subsets to analyse activated cells (Figure 4A). Table 2 lists the markers used to define the different memory T cell subsets.

qRT-PCR

After complete mechanical separation of the decidua basalis and parietalis, biopsies of 1cm x 1cm, without infarctions or calcifications, were taken from the central region of both layers. Biopsies were put in RNA Later (eBioscience, USA) and immediately stored at 4°C for 6 hours and were then put at -20°C until analysis. Tissue was disrupted with a TissueLyser (Qiagen, The Netherlands) (50 Hertz, 2 minutes) in 1% β-mercapto-ethanol (Sigma-Aldrich, USA) in RLT buffer (Qiagen, The Netherlands). RNA was isolated with an Allprep DNA/RNA mini kit (Qiagen, The Netherlands) following manufacturer-s instructions. Absorbance-based quantification of each RNA sample was performed using a NanoDrop Spectrophotometer (ND-1000, NanoDrop Technologies Inc, USA). Intactness of RNA samples was confirmed in random samples from both the decidua basalis and the decidua parietalis by electrophoresis in 2% agarose gel with ethidium bromide (0.2 µg). cDNA was transcribed with Super-script-II Reverse Transcriptase kit (Invitrogen, USA). Houskeeping genes

hypoxan-thine-guanine phosphoribosyltransferase (HPRT) (Hs02800695_m1) and β-actin

(10)

distributed and had a lower standard deviation (SD) compared to mRNA expression of ACTB in all decidua parietalis and decidua basalis samples. Therefore, HPRT was used as reference housekeeping gene for analysis56. mRNA expression of memory T cell associated cytokines IL7 (Hs00174202_m1), IL15 (Hs01003716_m1), and

IL23 (Hs00372324_m1), and preeclampsia associated cytokines interferon-γ (IFN-γ)

(Hs00989291_m1), interleukin-1β (IL1β) (Hs00174097_m1), IL2 (Hs00174114_m1),

IL6 (Hs00985639_m1), IL8 (Hs00174103_m1), and IL10 (Hs00961622_m1), was

analysed using Taqman Gene Expression Assays (Thermo Fisher, USA). PCR reactions were performed in triplicates with 15 ng cDNA in presence of Taqman Mastermix (Thermo Fisher, USA). Cycles were performed on a ViiA7-Real-Time PCR System (Thermo Fisher, USA). Data were normalized to housekeeping gene HPRT using 2−ΔCt.

Table 2. Memory T cell subsets and identification markers

  Memory T cell subset Markers used for identification

CD4+ Memory cells (CD4+) CD45RO+CD4+

Treg CD4+ memory Foxp3+CD45RO+CD4+

Central-Memory (CD4+ CM) CCR7+CD45RO+CD4+

Treg CD4+ CM cells Foxp3+CCR7+CD45RO+CD4+

Effector-Memory (CD4+ EM) CCR7-CD45RO+CD4+

Treg CD4+ EM cells Foxp3+CCR7-CD45RO+CD4+

Tissue Resident Memory (CD4+ TRM) CD103+CD45RO+CD4+

Treg CD4+ TRM Foxp3+CD103+CD45RO+CD4+

Activated CD4+ cell CD69+CD4+

CD8+ Memory cells (CD8+) CD45RO+CD8+

Central-Memory (CD8+ CM) CCR7+CD45RO+CD8+

Effector-Memory (CD8+ EM) CCR7-CD45RO+CD8+

  Tissue Resident Memory (CD8+ TRM) CD103+CD45RO+CD8+

Activated CD8+ cell CD69+CD8+

Central-Memory (CM), Effector-Memory (EM), T regulatory (Treg), Tissue Resident Memory (TRM), Forkhead box P3 (Foxp3), Chemokine receptor 7 (CCR7)

(11)

Statistics

Data were tested for normality using Kolmogorov-Smirnov test. When data were normally distributed, all groups were compared using one-way ANOVA followed by Tukey’s post hoc test. When data were distributed non-normally, the Kruskal-Wallis test followed by Dunn’s post hoc test was used to compare groups. Fisher’s exact test was used to analyse categorical variables. Depending on whether data was parametric or nonparametric, Pearson or Spearman correlation coefficient tests were used for correlation of mRNA expression with memory T cell proportions. Correlation coefficients of r ≥ 0.5 or r ≤ -0.5 were considered relevant. Linear regression analy-ses were performed to asanaly-sess the possible correlation between gestational age and memory T cell proportions. Outliers were excluded following the ROUT method57. Prism 7 software for Mac and Windows (GraphPad software Inc., USA; Microsoft, USA) and SPSS for Windows, version 23.0 software (SPSS Inc, Chicago, IL) was used. Differences were considered statistically significant if p < 0.05. A p-value < 0.10 was considered a statistical trend.

RESULTS

Patient characteristics

An overview of the patient characteristics is provided in table 1. Pregnancies complicated with LO-PE (p < 0.05) or EO-PE (p < 0.0001) had a lower gestational age compared to healthy pregnancies. The birth weight of babies after pregnancies complicated with EO-PE was lower compared to LO-PE (p < 0.0001) and healthy pregnancies (p < 0.0001), whereas babies after pregnancies complicated with LO-PE did not have a lower birth weight compared to healthy pregnancies. Systolic and diastolic blood pressure were significantly higher in EO-PE (p < 0.001) and LO-PE (p < 0.01) pregnancies compared to healthy pregnancies. In addition, more babies from EO-PE pregnancies were delivered through caesarean section compared to LO-PE pregnancies (p < 0.05). All women suffering from EO-PE were treated with corticosteroids for fetal lung maturation, whereas none of the women with LO-PE or healthy pregnancies did. No differences were found between the groups for any of the other characteristics.

(12)

Decidual CD4+ memory cell subsets in LO-PE, EO-PE and

healthy pregnancies

To investigate CD4+ memory T cell proportions at the fetal-maternal interface in preeclampsia, flow cytometric analyses were performed on lymphocytes isolated from the decidua parietalis and decidua basalis from pregnancies with LO-PE, EO-PE, and healthy pregnancies (Figure 1A-D, Supplementary Fig. S2).

In the decidua parietalis, the total CD4+ cell proportion of live lymphocytes did not significantly differ between the groups (not shown). Lower proportions of the general CD4+ memory cell (p < 0.01) population were found in LO-PE compared to healthy pregnancies in the decidua parietalis (Figure 1E). Lower CD4+ CM cell proportions were found in the decidua parietalis in EO-PE (p < 0.01) and even more in LO-PE (p < 0.0001) compared to healthy pregnancies (Figure 1F). A trend towards lower (p = 0.06) CD4+ TRM cells was found in EO-PE compared to healthy pregnancies in the decidua parietalis (Figure 1H). Lower Treg CD4+ CM cell proportions were found in the decidua parietalis from LO-PE compared to healthy pregnancies (p < 0.05) (Figure 1J). In the decidua parietalis, no differences were found for any of the other CD4+ memory cell populations between the groups. There were no significant differences in any of the CD4+ memory cell subsets between EO-PE and LO-PE.

Similarly to the decidua parietalis, in the decidua basalis, the total CD4+ cell pro-portion did not differ between the groups (not shown) and lower CD4+ CM cells were observed in LO-PE pregnancies compared to healthy pregnancies (p < 0.05) (Figure 1F). Apart from this CD4+ CM cell difference, in the decidua basalis no differences between the groups were found in any of the other CD4+ memory cell subsets, i.e. the general CD4+ memory, CD4+ EM, Treg CD4+ memory, Treg CD4+ CM, Treg CD4+ EM, CD4+ TRM, and Treg CD4+ TRM cell proportions (Figure 1). There were no signi-ficant differences in any of the CD4+ memory cell subsets between EO-PE and LO-PE.

Activated decidual CD4+ memory cell subsets in LO-PE, EO-PE and

healthy pregnancies

To determine the activation status of CD4+ memory cells, CD69 expression was analysed in all CD4+ memory cell subsets (Figure 2A)58.

In the decidua parietalis, a trend towards higher activated proportions of the general CD4+ memory cell population was found in EO-PE compared to healthy pregnancies

(13)

(p = 0.08) (Figure 2B). Higher activated proportions of the Treg CD4+ CM (p < 0.01), and the Treg CD4+ TRM (p < 0.05) cell populations were found in EO-PE pregnancies compared to healthy pregnancies (Figure 2G, 2I). When comparing the subtypes of preeclampsia in the decidua parietalis, the total CD4+ cell (p < 0.05) (not shown), the general CD4+ memory cell (p < 0.05), CD4+ CM cell (p < 0.01), CD4+ EM cell (p < 0.05), Treg CD4+ memory cell (p < 0.05), Treg CD4+ CM cell (p < 0.01), and

Figure 1. CD4+ memory cells in the decidua parietalis and the decidua basalis. Representative dot plots

showing fl ow cytometric analysis of decidua parietalis live CD4+ lymphocytes using forward/sideward

(FSC/SSC) scatterplots, viability stain and CD4 and CD8 expression (A). Within the CD4+ cell

popu-lation, CD4+ central-memory (CM) cells (CCR7+CD45RO+CD4+) (B), CD4+ effector-memory (EM) cells

(CCR7-CD45RO+CD4+) (B), general CD4+ memory cells (CM+EM) (CD45RO+CD4+) (B), and CD4+ tissue

resident memory (TRM) cells (CD103+CD45RO+CD4+) (C) were identifi ed. Within the general CD4+

me-mory cell population, T regulatory (Treg) CD4+ memory cells (Foxp3+CD45RO+CD4+) were identifi ed

(D). Proportions of general CD4+ memory cells (E), CD4+ CM cells (F), CD4+ EM cells (G), CD4+ TRM

cells (H), Treg CD4+ memory cells (I), Treg CD4+ CM (Foxp3+CCR7+CD45RO+CD4+) cells (J), Treg CD4+

EM (Foxp3+CCR7-CD45RO+CD4+) cells (K), and Treg CD4+ TRM (Foxp3+CD103+CD45RO+CD4+) cells

(L), in the decidua parietalis and the decidua basalis from healthy pregnancies and pregnancies

com-plicated by late-onset preeclampsia (LO-PE) or early-onset preeclampsia (EO-PE), shown as proportion

of the CD4+ cell population. Symbols represent individual values per decidua with data as median with

interquartile range. Analysis by Kolmogorov-Smirnov test, one-way ANOVA and Tukey’s post hoc test or Kruskal- Wallis and Dunn’s post hoc test comparing the different groups within the decidua parietalis or the decidua basalis; *p < 0.05, **p < 0.01, **** p < 0.0001.

(14)

Treg CD4+ TRM cell (p < 0.05) populations, showed a higher activated proportion in EO-PE pregnancies compared to LO-PE pregnancies (Figure 2B-2G, 2I).

In the decidua basalis, lower activated proportions of the general CD4+ memory cell population (p < 0.05) and the CD4+ EM cell population (p < 0.01) were found in LO-PE compared to healthy pregnancies (Figure 2B, 2D). In the CD4+ CM cell population (p < 0.01) and the TRM CD4+ memory cell population (p < 0.05), lower

Figure 2. Activated CD4+ memory cells in the decidua parietalis and the decidua basalis. Representative

dot plots showing fl ow cytometric analysis of decidua parietalis activated (CD69+) general CD4+ memory

cells (CM+EM) (CD69+CD45RO+CD4+), CD4+ central-memory (CM) cells (CD69+CCR7+CD45RO+CD4+),

CD4+ effector-memory (EM) cells (CD69+CCR7-CD45RO+CD4+), CD4+ tissue resident memory (TRM) cells

(CD69+CD103+CD45RO+CD4+), and T regulatory (Treg) CD4+ memory cells (CD69+Foxp3+CD45RO+

-CD4+) (A). Proportions of activated cells of the CD4+ memory cell population (B), the CD4+ CM cell

population (C), the CD4+ EM cell population (D), the CD4+ TRM cell population (E), the Treg CD4+

memory cell population (F), the Treg CD4+ CM (Foxp3+CCR7+CD45RO+CD4+) cell population (G),

the Treg CD4+ EM (Foxp3+CCR7-CD45RO+CD4+) cell population (H), and the Treg CD4+ TRM

(Fox-p3+CD103+CD45RO+CD4+) cell population (I), in the decidua parietalis and the decidua basalis from

healthy pregnancies and pregnancies complicated by late-onset preeclampsia (LO-PE) or early-onset preeclampsia (EO-PE). Symbols represent individual values per decidua with data as median with in-terquartile range. Analysis by Kolmogorov-Smirnov test, one-way ANOVA and Tukey’s post hoc test or Kruskal-Wallis and Dunn’s post hoc test comparing the different groups within the decidua parietalis or the decidua basalis; *p < 0.05, **p < 0.01.

(15)

activated proportions were found in the decidua basalis from both preeclamptic groups compared to the control group (Figure 2C, 2E). No differences in activation in the total CD4+ cell population and in any of the memory T cell subsets were found between the preeclampsia subtypes in the decidua basalis.

Comparing the decidua parietalis and basalis, lower activated proportions of all CD4+ memory cell subsets were found in EO-PE in the decidua basalis compared to the decidua parietalis (p < 0.05) (Figure 2). The activated proportions did not differ between the two layers in healthy and LO-PE pregnancies.

Decidual CD8+ memory cell subsets in LO-PE, EO-PE and

healthy pregnancies

The general CD8+ memory cell population, the CD8+ CM cell population, the CD8+ EM cell population, and the CD8+ TRM cell population were analysed by flow cytometry in the decidua basalis and decidua parietalis from LO-PE, EO-PE and healthy pregnancies (Figure 3A, 3B, Supplementary Fig. S3). We did not find any Foxp3+CD8+ memory cells in the decidual layers.

In the decidua parietalis, the total CD8+ cell proportion of live lymphocytes did not significantly differ between the groups (not shown). The general CD8+ memory cell population (p < 0.05) and the CD8+ CM cell population (p < 0.01) were lower in EO-PE compared to healthy pregnancies in the decidua parietalis (Figure 3C, 3D). The general CD8+ memory cell population was also lower in LO-PE compared to healthy pregnancies in the decidua parietalis (p < 0.05) (Figure 3C). CD8+ EM cells showed a trend towards lower proportions in EO-PE compared to healthy pregnan-cies in the decidua parietalis (p = 0.06) (Figure 3E). No differences were observed between the groups in CD8+ TRM cell proportions in the decidua parietalis (Figure 3F). The preeclampsia subtypes had no effect on CD8+ memory cell proportions in the decidua parietalis.

In the decidua basalis, the general CD8+ memory cell proportion was lower in EO-PE compared to healthy pregnancies (p < 0.05) (Figure 3C). No differences in the total CD8+ cell population and in any of the other CD8+ memory cell subsets were found between the groups in the decidua basalis.

(16)

Figure 3. CD8+ memory cells in the decidua parietalis and the decidua

basa-lis. Representative dot plots showing fl ow cytometric analysis of decidua

pa-rietalis CD8+ central-memory (CM) cells (CCR7+CD45RO+CD8+) (A), CD8+

effector-memory (EM) cells (CCR7-CD45RO+CD8+) (A), general CD8+ memory

cells (CM+EM) (CD45RO+CD8+) (A), and CD8+ tissue resident memory (TRM) cells

(CD103+CD45RO+CD8+) (B), identifi ed in the CD8+ cell population. Proportions

of general CD8+ memory cells (C), CD8+ CM cells (D), CD8+ EM cells (E), and

CD8+ TRM cells (F), in the decidua parietalis and the decidua basalis from healthy

pregnancies and pregnancies complicated by late-onset preeclampsia (LO-PE) or

early-onset preeclampsia (EO-PE), shown as proportion of the CD8+ cell

popula-tion. Symbols represent individual values per decidua with data as median with interquartile range. Analysis by Kolmogorov-Smirnov test, one-way ANOVA and Tukey’s post hoc test or Kruskal-Wallis and Dunn’s post hoc test comparing the different groups within the decidua parietalis or the decidua basalis; *p < 0.05, **p < 0.01.

(17)

Figure 4. Activated CD8+ memory cells in the decidua parietalis and the decidua

ba-salis. Representative dot plots showing fl ow cytometric analysis of decidua parietalis

activated (CD69+) general CD8+ memory cells (CM+EM) (CD69+CD45RO+CD8+),

CD8+ central-memory (CM) cells (CD69+CCR7+CD45RO+CD8+), CD8+

effec-tor-memory (EM) cells (CD69+CCR7-CD45RO+CD8+), and CD8+ tissue resident

memory (TRM) cells (CD69+CD103+CD45RO+CD8+) (A). Proportions of activated

cells of the CD8+ memory cell population (B), the CD8+ CM cell population (C),

the CD8+ EM cell population (D), the CD8+ TRM cell population (E), in the decidua

parietalis and the decidua basalis from healthy pregnancies and pregnancies complicated by late-onset preeclampsia (LO-PE) or early-onset preeclampsia (EO-PE). Symbols represent individual values per decidua with data as median with interquartile range. Analysis by Kolmogorov-Smirnov test, one-way ANOVA and Tukey’s post hoc test or Kruskal-Wallis and Dunn’s post hoc test comparing the different groups within the decidua parietalis or the decidua basalis; *p < 0.05, **p < 0.01.

(18)

Activated decidual CD8+ memory cell subsets in LO-PE, EO-PE and

healthy pregnancies

CD69 expression was used to analyse the activated proportions of CD8+ memory cells (Figure 4A)58.

In the decidua parietalis a trend towards higher activated (CD69+) proportions of the general CD8+ memory cell population (p = 0.07), the CD8+ CM cell population (p = 0.06), the CD8+ EM cell population (p = 0.09), and a significantly higher pro-portion of the CD8+ TRM cell population (p < 0.05) was observed in EO-PE compared to healthy pregnancies (Figure 4B-4E). When comparing the preeclampsia subtypes, higher activated proportions of the total CD8+ cell population (not shown), and all CD8+ memory cell subsets were found in EO-PE compared to LO-PE in the decidua parietalis (p < 0.05) (Figure 4B-4E). This was similar to the activation of the memory T cell subsets in the CD4+ cell lineage in the decidua parietalis (Figure 2).

In the decidua basalis, lower activated proportions of the general CD8+ memory cell population (p < 0.05), and a trend towards lower activated proportions of the CD8+ EM cell population (p = 0.07) and CD8+ TRM cell population (p = 0.06) was found in LO-PE compared to healthy pregnancies (Figure 4B, 4D, 4E). Activated proportions of the CD8+ CM cell population did not differ between the groups in the decidua basalis (Figure 4C). Comparison of the preeclampsia subtypes showed no difference in activated proportions of the total CD8+ cell population (not shown) and higher activated proportions of general CD8+ memory cells (p < 0.01), CD8+ EM cells (p < 0.01) and CD8+ TRM cells (p < 0.05) in EO-PE pregnancies compared to LO-PE pregnancies (Figure 4B, 4D, 4E).

Lower activated proportions of all CD8+ memory cell subsets were found in EO-PE in the decidua basalis compared to the decidua parietalis (p < 0.05) (Figure 4). The activated proportions did not differ between the two layers in healthy and LO-PE pregnancies.

mRNA expression of memory T cell associated cytokines in biopsies from the decidua parietalis and basalis in LO-PE, EO-PE and

healthy pregnancies

To explore pathways that could contribute to the lower memory T cell populations in preeclampsia, qRT-PCR analysis was performed on biopsies from the decidua

(19)

basalis and decidua parietalis for analysis of mRNA expression of IL7, IL15, and

IL23 (Figure 5).

In the decidua parietalis, a trend towards higher IL15 mRNA expression was found in EO-PE pregnancies compared to healthy pregnancies (p = 0.07) (Figure 5B). In the decidua parietalis, no differences were found between the groups for IL7 and

IL23 mRNA expression (Figure 5A, 5C). The preeclampsia subtypes did not show

any effect on IL7, IL15, and IL23 mRNA expression in the decidua parietalis. In the decidua basalis, IL7 mRNA expression was lower in EO-PE pregnancies as compared to healthy pregnancies (p < 0.05) (Figure 5A). mRNA expression of

IL15 (p < 0.05) and IL23 (p < 0.05) was higher in the decidua basalis from EO-PE

pregnancies compared to healthy pregnancies (Figure 5B, 5C). Also in the decidua basalis, a trend towards higher mRNA expression of IL15 (p = 0.09) and signifi cantly higher mRNA expression of IL23 (p < 0.05) was found in LO-PE compared to healthy pregnancies (Figure 5B, 5C). Comparison of the preeclampsia subtypes showed no differences in mRNA expression of IL7, IL15, and IL23 in the decidua basalis.

Correlation analyses in the decidua parietalis showed that mRNA expression of

IL15 was associated with activated proportions of CD8+ memory cells (p < 0.05, r = 0.50) and CD8+ CM cells (p < 0.01, r = 0.54), whereas in the decidua basalis no association with memory T cell proportions was found (Supplementary Tables S1 and Figure 5. mRNA expression of memory T cell associated cytokines in the decidua. mRNA expression of IL7 (A), IL15 (B), and IL23 (C), in biopsies from the decidua parietalis and the decidua basalis from healthy pregnancies and pregnancies complicated by late-onset preeclampsia (LO-PE) or early-onset preeclampsia (EO-PE). Data are shown as median with interquartile range mRNA target gene expression normalized to housekeeper gene HPRT. Analysis by Kolmogorov-Smirnov test, one-way ANOVA and Tukey’s post hoc test or Kruskal-Wallis and Dunn’s post hoc test comparing the different groups within the decidua parietalis or the decidua basalis; *p < 0.05.

(20)

S2). mRNA expression of IL7 and IL23 were not associated with any of the memory T cell proportions in either the decidua parietalis or basalis.

Figure 6. mRNA expression of preeclampsia associated cytokines in the decidua. mRNA expression of interferon-γ (IFN-γ) (A), IL1β (B), IL2 (C), IL6 (D), IL8 (E), and IL10 (F), in biopsies from the decidua parie-talis and the decidua basalis from healthy pregnancies and pregnancies complicated by late-onset pree-clampsia (LO-PE) or early-onset preepree-clampsia (EO-PE). Data are shown as median with interquartile range mRNA target gene expression normalized to housekeeper gene HPRT. Analysis by Kolmogorov-Smirnov test, one-way ANOVA and Tukey’s post hoc test or Kruskal-Wallis and Dunn’s post hoc test comparing the different groups within the decidua parietalis or the decidua basalis; *p < 0.05, **p < 0.01.

mRNA expression of preeclampsia associated cytokines in biopsies from the decidua parietalis and basalis in LO-PE, EO-PE and

healthy pregnancies

To gain insight into the infl ammatory cytokine milieu that could be affected by, or affect, memory T cells in preeclampsia, mRNA expression of preeclampsia associa-ted cytokines, IFN-γ, IL1β, IL2, IL6, IL8 and IL10 was analysed in biopsies from the decidua parietalis and decidua basalis from LO-PE, EO-PE, and healthy pregnancies (Figure 6)59,60.

In the decidua parietalis, IFN-γ showed a trend towards higher mRNA expression in EO-PE compared to healthy pregnancies (p = 0.06) (Figure 6A). Higher mRNA expression of IL2 (p < 0.01) was found in the decidua parietalis from EO-PE compared to healthy pregnancies (Figure 6C). No differences in mRNA expression of IL1β were found between the groups in the decidua parietalis (Figure 6B). Comparison of the

(21)

preeclampsia subtypes showed a trend towards higher IFN-γ (p = 0.09) and IL2 (p = 0.07) mRNA expression and a trend towards lower IL6 (p = 0.10) and IL8 (p = 0.10) mRNA expression in EO-PE compared to LO-PE in the decidua parietalis (Figure 6A, 6C-6E).

In the decidua basalis, higher mRNA expression of IL1β in LO-PE (p < 0.05) and a trend towards higher expression in EO-PE (p = 0.07) was found compared to healthy pregnancies (Figure 6B). Analysis of IL2, IL6, IL8, and IL10 mRNA expression did not show any significant differences between the groups in the decidua basalis (Figure 6C-6F). When comparing the effect of the preeclampsia subtypes on mRNA expression of preeclampsia associated cytokines in the decidua basalis a trend towards lower

IFN-γ was observed in EO-PE compared to LO-PE (p = 0.09) (Figure 6A).

Correlation analyses in the decidua parietalis showed that mRNA expression of

IFN-γ was associated with activated proportions of CD8+ memory cells (p < 0.01, r = 0.62), CD8+ CM cells (p < 0.01, r = 0.59), and CD8+ TRM cells (p < 0.001, r = 0.67), and that mRNA expression of IL1β was associated with CD4+ CM cells (p < 0.05, r = -0.51). mRNA expression of IL2 was associated with activated propor-tions of all CD8+ memory cell subsets (p < 0.01-0.05, r = 0.51-0.58), and IL8 was associated with activated proportions of CD4+ memory cells (p < 0.05, r = -0.52), CD4+ EM cells (p < 0.001, r = -0.67), CD8+ memory cells (p < 0.01, r = -0.54), and CD8+ TRM cells (p < 0.01, r = -0.54) (Supplementary Table S1). In the decidua basalis, IFN-γ mRNA expression was associated with CD4+ EM cells (p < 0.01, r = 0.51), CD4+ TRM cells (p < 0.01, r = 0.56), CD8+ TRM cells (p < 0.01, r = 0.53), and activated proportions of CD8+ CM cells (p < 0.05, r = 0.50) (Supplementary Table S2). No association was found for mRNA expression for any of the other genes in the decidua basalis.

DISCUSSION

In this study, we showed fewer CD4+ CM cells and CD8+ memory cells in the decidua parietalis from EO-PE and LO-PE compared to healthy pregnancies. All CD4+ and CD8+ memory cell subsets analysed in this study had higher activated proportions in the decidua parietalis from EO-PE compared to LO-PE. These findings were accom-panied by lower IL7 mRNA expression, a memory T cell associated cytokine, and higher mRNA expression of IL15 and IL23 in the decidua basalis in EO-PE compared to healthy pregnancies.

(22)

In accordance with previous studies, our study showed that various memory T cell subpopulations are present at the fetal-maternal interface16,20. Since our methods were designed to minimize contamination of decidual T cells with fetal T cells and T cells from maternal blood (see Supplementary Fig. S4 and Supplementary Methods), we consider the memory T cells analysed in this study, a good representation of the decidual memory T cell populations. Whether these cells play a role in fetal-maternal immune tolerance at the fetal-maternal interface remains unknown. Based on their well-known function in transplant rejection61, one may wonder why these cells are present at the fetal-maternal interface. Previously, Tilburgs et al. showed that CD8+ memory cells at the fetal-maternal interface have decreased cytotoxic activity, as they showed reduced levels of perforin and granzyme B, but increased levels of programmed cell death-1 (PD-1)3. In contrast with this study, other studies showed that decidual CD8+ EM cells have highly upregulated genes associated with immune effector processes and are still functional upon stimulation, showing high capacity to proliferate, degranulate, and produce cytokines43,44,62. Therefore, the function of T cells, especially memory T cells, at the fetal-maternal interface requires further investigation. Indeed, mice studies suggest that the function of memory T cells during pregnancy has changed1,41,42. It may be suggested that memory T cells are differently regulated at the fetal-maternal interface, which affects their function and keeps them quiescent throughout pregnancy to generate tolerance to fetal antigens, however remain capable of eliciting an immune response towards pathogens.

Our finding of lower proportions of CD4+ CM and CD8+ memory cells, in decidua parietalis tissue from preeclamptic pregnancies compared to healthy pregnancies, is in line with the latter suggestion of a role of memory T cells in fetal tolerance at the fetal-maternal interface. Our findings suggest that CM and EM cells are differently maintained at the fetal maternal interface. Indeed, CM and EM cells have different homing capacities and homing receptors23,31,32. CM cells are mainly thought to provide central immune surveillance by patrolling the lymph nodes draining peripheral tissues, while EM cells are mainly thought to migrate to non-lymphoid tissue31. However, our study and various other studies have shown that the distinction between CM and EM cells may not be so clear, since CM cells are also found in non-lymphoid tissue and in these tissues may have effector functions similar to EM cells3,44,63. CD4+ CM cells are long-living cells and are able to persist in the circulation for a long time64. They can also transform into different CD4+ cell subsets, including EM cells64,65. It may be that CD4+ CM cells persist in the circulation after pregnancy. This suggestion is in line with our previous study in parous women after uncomplicated pregnancies, which showed persisting higher levels of CD4+ CM cells in women postpartum compared

(23)

to nulligravid women66. If these persistent CD4+ CM cell populations are lower in women after a preeclamptic pregnancy compared to women after a healthy preg-nancy is under investigation in our lab. If and how these memory T cell populations would induce paternal antigen specific tolerance instead of immunity against paternal antigens remains to be investigated.

In the present study, we found CD4+ Treg memory cells in the decidua parietalis and the decidua basalis in both healthy and preeclamptic pregnancies. We did not find CD8+ memory cells expressing Foxp3 in the decidua. This is in line with suggestions that Treg CD8+ memory cells do exist, but are distinguished by other markers39,40. Within the CD4+ cell population, we were able to identify Treg CM, Treg EM and Treg TRM cells. As Rosenblum et al. suggested in their review, this implies that within the Treg CD4+ memory cell populations, similar subpopulations exist as compared with the CD4+ memory cell subsets28. Since studies in mice have shown a Treg CD4+ memory cell population with fetal antigen specificity which is generated during gestation, remains postpartum and contributes to lower resorption rates in subsequent pregnancies1, we expected decreased proportions of these cells in the decidua of women with preeclampsia. Indeed, we observed lower Treg CD4+ CM cells in the decidua parietalis from LO-PE compared to healthy pregnancies. However, other Treg CD4+ memory cell populations did not differ between healthy and pree-clamptic pregnancies. Our study may thus indicate that Treg CD4+ memory cells in the decidua are not so much affected in preeclampsia. As it is known that Treg cells are increased at mid gestation and decrease towards the end of pregnancy67,68, the lack of a decrease of Treg CD4+ memory cells in preeclampsia may also be due to differences in gestational age between the groups, especially between the healthy pregnancy group and the EO-PE group, which have a large difference in gestational age. As this difference is insurmountable but could affect our results, we performed linear regression analyses in the healthy pregnancy group separately and in all groups together. The analyses did not show any significant effect of gestational age on memory T cell proportions. Although the sample size of our study is limited, the very homogenous patient groups are a strength. In this study, we prioritized homo-genous patient groups over a bigger cohort, given the multifactorial character of the pathophysiology of preeclampsia.

To explore potential mechanisms related to decreased memory T cell populations in the preeclamptic decidua, we measured mRNA expression of various memory T cell associated cytokines in biopsies from the decidua parietalis and basalis. We showed slightly lower IL7 mRNA expression in the decidua basalis in LO-PE and significantly

(24)

lower IL7 mRNA expression in EO-PE compared to healthy pregnancies. IL7 promotes survival and self-renewal of both CD4+ and CD8+ memory cell lineages69,70, which could indicate that the decreased proportions of CD8+ memory T cell populations in the preeclamptic decidua basalis are due to decreased levels of IL7, however correlation analyses did not show a relevant association. IL15 and IL23 are pro-inflammatory cytokines that have stimulatory effects on CD4+ and CD8+ memory cells, promote long term survival, and induce IFN-γ, tumour necrosis factor B, and IL17 secretion25,71–73. The higher levels of IL15 and IL23 mRNA expression in the decidua basalis from preeclamptic pregnancies in the face of lower levels of memory T cell populations, indicate that these cytokines may not be involved in the decreased proportions of memory T cells in the decidua. The levels of IL15 in the decidua parietalis might be involved in the higher activated proportions of CD8+ memory cell subsets, since, the

IL15 mRNA expression was associated with the higher activated proportions of CD8+ memory cells, CD8+ CM cells, and CD8+ TRM cells. It should be noted that mRNA expression analyses were performed on tissue biopsies, therefore mRNA expression levels reflect the general cytokine environment produced by all immune cells present in the biopsy and not memory T cells specifically.

Although several findings were similar in both the decidua basalis and decidua parietalis, many were specific for one of the decidual layers. Various papers have shown that the decidua parietalis and basalis have different functional characteris-tics and consist of a different immune cell repertoire possibly due to differential HLA expression on trophoblasts and prostaglandin secretion in the different anatomic loca-tions15–18,74. The most prominent difference between the two decidual compartments in this study are the lower activated proportions of all CD4+ and CD8+ memory cell subsets in EO-PE in the decidua basalis compared to the decidua parietalis. This might be due to the cytokines present in the decidua parietalis since correlation analyses showed that activated proportions of the memory T cells were associated with IL8,

IL15, and IFN-γ mRNA expression in the decidua parietalis and not in the decidua

basalis. Distinguishing the different decidual layers is a strength from our study, which differs from the study of Nguyen et al. who showed a shift from naive to memory T cell phenotype in preeclampsia4. These authors did not analyse the decidua, but instead used lymphocytes from a swap of the intrauterine cavity4.

In this study we distinguished between LO-PE and EO-PE as both subtypes of pree-clampsia have a different pathophysiology9,10. When we compared EO-PE and LO-PE we found higher activated proportions in almost every memory T cell subset analysed in the decidua parietalis (apart from CD4+ EM cells, p = 0.07) from EO-PE compared

(25)

to LO-PE. mRNA expression of preeclampsia associated cytokines showed a trend towards higher mRNA expression of IFN-γ and IL2 and lower mRNA expression of

IL6 and IL8 in the decidua parietalis from EO-PE compared to LO-PE. Since EO-PE is

associated with higher oxidative stress compared to LO-PE9,10,75, and oxidative stress is associated with higher CD69 expression76, the higher activated proportions of memory T cells in EO-PE compared to LO-PE in this study may be due to higher oxida-tive stress. Also, excess syncytiotrophoblast microparticle shedding into the maternal circulation in EO-PE and not LO-PE could cause increased activated proportions and higher IFN-γ and IL2 mRNA expression in EO-PE compared to LO-PE77. It needs to be indicated that an effect of gestational age between EO-PE and LO-PE may also be involved in the differences between the subtypes of preeclampsia.

In conclusion, this study showed that memory T cell subsets in the decidua basalis and parietalis are altered in EO-PE and LO-PE compared to healthy pregnancies. In addition, higher activated proportions of memory T cells were found in EO-PE com-pared to LO-PE in the decidua parietalis. These findings were accompanied by lower mRNA expression of the memory T cell stimulating cytokine IL7 and higher IL15 and

IL23 mRNA expression in EO-PE compared to healthy pregnancies. We hypothesize

that the decreased presence of specific memory T cell subsets at the fetal-maternal interface in preeclampsia may play a role in the pathophysiology of preeclampsia, i.e. decreased fetal tolerance. Further studies into the function of memory T cells at the fetal-maternal interface should shed light on their exact function in fetal-mater-nal tolerance.

(26)

REFERENCES

1. Rowe, J. H., Ertelt, J. M., Xin, L. & Way, S. S. Pregnancy imprints regulatory memory

that sustains anergy to fetal antigen. Nature 490, 102–6 (2012).

2. Kinder, J. M. et al. Cross-Generational Reproductive Fitness Enforced by Microchimeric

Maternal Cells. Cell 162, 505–515 (2015).

3. Tilburgs, T. et al. Human decidual tissue contains differentiated CD8+ effector-memory

T cells with unique properties. J. Immunol. 185, 4470–7 (2010).

4. Nguyen, T. A., Kahn, D. A. & Loewendorf, A. I. Maternal—Fetal rejection reactions are

unconstrained in preeclamptic women. PLoS One 12, e0188250 (2017).

5. Saftlas, A. F., Olson, D. R., Franks, A. L., Atrash, H. K. & Pokras, R. Epidemiology of

preeclampsia and eclampsia in the United States, 1979-1986. Am. J. Obstet. Gynecol.

163, 460–5 (1990).

6. Abalos, E., Cuesta, C., Grosso, A. L., Chou, D. & Say, L. Global and regional estimates

of preeclampsia and eclampsia: a systematic review. Eur. J. Obstet. Gynecol. Reprod. Biol. 170, 1–7 (2013).

7. Steegers, E. A. P., von Dadelszen, P., Duvekot, J. J. & Pijnenborg, R. Pre-eclampsia.

Lancet 376, 631–44 (2010).

8. Tranquilli, A. L., Brown, M. A., Zeeman, G. G., Dekker, G. & Sibai, B. M. The definition

of severe and early-onset preeclampsia. Statements from the International Society for the Study of Hypertension in Pregnancy (ISSHP). Pregnancy Hypertens. An Int. J. Women’s Cardiovasc. Heal. 3, 44–47 (2013).

9. Staff, A. C. & Redman, C. W. G. The Differences Between Early- and Late-Onset

Pre-eclampsia. in Preeclampsia, Comprehensive Gynecology and Obstetrics (ed. Saito, S.) 157–172 (Springer Nature Singapore Pte Ltd. 2018, 2018).

10. Raymond, D. & Peterson, E. A Critical Review of Early-Onset and Late-Onset Preeclampsia. Obstet. Gynecol. Surv. 66, 497–506 (2011).

11. Zenclussen, A. C., Schumacher, A., Zenclussen, M. L., Wafula, P. & Volk, H. D. Immunology of pregnancy: cellular mechanisms allowing fetal survival within the maternal uterus. Expert Rev. Mol. Med. 9, 1–14 (2007).

12. Prins, J. R. et al. Preeclampsia is associated with lower percentages of regulatory T cells in maternal blood. Hypertens. Pregnancy 28, 300–311 (2009).

13. Cudihy, D. & Lee, R. The pathophysiology of pre-eclampsia: current clinical concepts. J. Obstet. Gynaecol. 29, 576–582 (2009).

14. Saito Masatoshi, S. S., Saito, S. & Sakai, M. Th1/Th2 balance in preeclampsia. J. Reprod. Immunol. 59, 161–173 (2003).

15. Baergen, R. N. Histology of the Chorionic Villi, Fetal Membranes, and Umbilical Cord. in Manual of Benirschke and Kaufmann’s 80–95 (Springer-Verlag, 2005).

16. Tilburgs, T. et al. Differential Distribution of CD4+CD25bright and CD8+CD28- T-cells in Decidua and Maternal Blood During Human Pregnancy. Placenta 27, 47–53 (2006). 17. Baergen, R. N. Development and Histology of the Nonvillous Portions of the Placenta.

in Manual of Benirschke and Kaufmann’s 107–134 (Springer-Verlag, 2005).

18. Schonkeren, D. et al. Differential Distribution and Phenotype of Decidual Macrophages in Preeclamptic versus Control Pregnancies. Am. J. Pathol. 178, 709–717 (2011). 19. Guerin, L. R., Prins, J. R. & Robertson, S. A. Regulatory T-cells and immune tolerance

in pregnancy: a new target for infertility treatment? Hum. Reprod. Update 15, 517–535 (2009).

20. Nancy, P. & Erlebacher, A. T cell behavior at the maternal-fetal interface. Int. J. Dev. Biol. 58, 189–98 (2014).

21. Vento-Tormo, R. et al. Single-cell reconstruction of the early maternal–fetal interface in humans. Nature 563, 347–353 (2018).

(27)

22. Kieffer, T. E. C., Laskewitz, A., Scherjon, S. A., Faas, M. M. & Prins, J. R. Memory T Cells in Pregnancy. Front. Immunol. 10, 625 (2019).

23. Sallusto, F., Lenig, D., Förster, R., Lipp, M. & Lanzavecchia, A. A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature

401, 708–712 (1999).

24. Lanzavecchia Federica, A. S. Understanding the generation and function of memory T cell subsets. Curr. Opin. Immunol. 17, 326–332 (2005).

25. Li, Y., Wang, H., Lu, H. & Hua, S. Regulation of Memory T Cells by Interleukin-23. Int. Arch. Allergy Immunol. 169, 157–62 (2016).

26. Kondrack, R. M. et al. Interleukin 7 Regulates the Survival and Generation of Memory CD4 Cells. J. Exp. Med. 198, 1797–1806 (2003).

27. Richer, M. J. et al. Inflammatory IL-15 is required for optimal memory T cell responses. J. Clin. Invest. 125, 3477–90 (2015).

28. Rosenblum, M. D., Way, S. S. & Abbas, A. K. Regulatory T cell memory. Nat. Rev. Immunol. 1–12 (2015).

29. Topham, D. J. & Reilly, E. C. Tissue-Resident Memory CD8+ T Cells: From Phenotype to

Function. Front. Immunol. 9, 515 (2018).

30. Sallusto, F. et al. Switch in chemokine receptor expression upon TCR stimulation reveals novel homing potential for recently activated T cells. Eur. J. Immunol.

29, 2037–2045 (1999).

31. Sallusto, F., Geginat, J. & Lanzavecchia, A. Central memory and effector memory T cell subsets: function, generation, and maintenance. Annu. Rev. Immunol.

22, 745–63 (2004).

32. Förster, R. et al. CCR7 coordinates the primary immune response by establishing functional microenvironments in secondary lymphoid organs. Cell 99, 23–33 (1999). 33. Kumar, B. V. et al. Human Tissue-Resident Memory T Cells Are Defined by Core

Transcriptional and Functional Signatures in Lymphoid and Mucosal Sites. Cell Rep.

20, 2921–2934 (2017).

34. Chen, T. et al. Self-specific memory regulatory T cells protect embryos at implantation in mice. J. Immunol. 191, 2273–2281 (2013).

35. Kalekar, L. A. et al. CD4+ T cell anergy prevents autoimmunity and generates regulatory

T cell precursors. Nat. Immunol. 17, 304–314 (2016).

36. Tilburgs, T. et al. Evidence for a selective migration of fetus-specific CD4+CD25bright

regulatory T cells from the peripheral blood to the decidua in human pregnancy. J. Immunol. 180, 5737–5745 (2008).

37. Kälble, F. et al. Aberrant ICOS + -T cell differentiation in women with spontaneous

preterm labor. Am. J. Reprod. Immunol. 76, 415–425 (2016).

38. Schober, L. et al. The role of regulatory T cell (Treg) subsets in gestational diabetes mellitus. Clin. Exp. Immunol. 177, 76–85 (2014).

39. Wang, S.-C. S. et al. PD-1 and Tim-3 pathways are associated with regulatory CD8+

T-cell function in decidua and maintenance of normal pregnancy. Cell Death Dis.

6, e1738–e1738 (2015).

40. Machicote, A., Belén, S., Baz, P., Billordo, L. A. & Fainboim, L. Human CD8+HLA-DR+

Regulatory T Cells, Similarly to Classical CD4+Foxp3+ Cells, Suppress Immune Responses

via PD-1/PD-L1 Axis. Front. Immunol. 9, 2788 (2018).

41. Barton, B. M., Xu, R., Wherry, E. J. & Porrett, P. M. Pregnancy promotes tolerance to future offspring by programming selective dysfunction in long-lived maternal T cells. J. Leukoc. Biol 101, 975–987 (2017).

42. Kahn, D. A. & Baltimore, D. Pregnancy induces a fetal antigen-specific maternal T regulatory cell response that contributes to tolerance. Proc. Natl. Acad. Sci. U. S. A. 107, 9299–304 (2010).

(28)

43. van der Zwan, A. et al. Mixed signature of activation and dysfunction allows human

decidual CD8+ T cells to provide both tolerance and immunity. Proc. Natl. Acad. Sci.

U. S. A. 115, 385–390 (2018).

44. Powell, R. M. et al. Decidual T Cells Exhibit a Highly Differentiated Phenotype and Demonstrate Potential Fetal Specificity and a Strong Transcriptional Response to IFN. J. Immunol. 199, 3406–3417 (2017).

45. Zeng, W. et al. Distinct transcriptional and alternative splicing signatures of decidual

CD4+ T cells in early human pregnancy. Front. Immunol. 8, 682 (2017).

46. Wagner, M. I. et al. Differentiation of ICOS+ and ICOS- recent thymic emigrant

regulatory T cells (RTE T regs) during normal pregnancy, pre-eclampsia and HELLP syndrome. Clin. Exp. Immunol. 183, 129–42 (2016).

47. Wagner, M. I. et al. The role of recent thymic emigrant-regulatory T-cell (RTE-Treg) differentiation during pregnancy. Immunol. Cell Biol. 93, 858–867 (2015).

48. Robillard, P. Y. et al. Paternity patterns and risk of preeclampsia in the last pregnancy in multiparae. J. Reprod. Immunol. 24, 1–12 (1993).

49. Tubbergen, P. et al. Change in paternity: a risk factor for preeclampsia in multiparous women? J. Reprod. Immunol. 45, 81–8 (1999).

50. Li, D. K. & Wi, S. Changing paternity and the risk of preeclampsia/eclampsia in the subsequent pregnancy. Am. J. Epidemiol. 151, 57–62 (2000).

51. Saftlas, A. F. et al. Cumulative exposure to paternal seminal fluid prior to conception and subsequent risk of preeclampsia. J. Reprod. Immunol. 101–102, 104–110 (2014). 52. Robillard, P. Y. et al. Association of pregnancy-induced hypertension with duration of

sexual cohabitation before conception. Lancet 344, 973–5 (1994).

53. Brown, M. A. et al. Hypertensive Disorders of Pregnancy: ISSHP Classification, Diagnosis, and Management Recommendations for International Practice. Hypertension

72, 24–43 (2018).

54. Committee for Guidelines in Research (COREON). Human Tissue and Medical Research: Code of Conduct for responsible use (2011). (2011).

55. van Egmond, A. et al. Preservation of human placenta is feasible and facilitates studies on the local immune regulation in normal and aberrant pregnancies. J. Reprod. Immunol.

94, 45 (2012).

56. Mane, V. P., Heuer, M. A., Hillyer, P., Navarro, M. B. & Rabin, R. L. Systematic method for determining an ideal housekeeping gene for real-time PCR analysis. J. Biomol. Tech.

19, 342–7 (2008).

57. Motulsky, H. J. et al. Detecting outliers when fitting data with nonlinear regression – a new method based on robust nonlinear regression and the false discovery rate. BMC Bioinformatics 7, 123 (2006).

58. Ziegler F Alderson, M R., S. F. R., Ziegler, S. F., Ramsdell, F. & Alderson, M. R. The activation antigen CD69. Stem Cells 12, 456–465 (1994).

59. Pinheiro, M. B. et al. Severe preeclampsia goes along with a cytokine network disturbance towards a systemic inflammatory state. Cytokine 62, 165–173 (2013). 60. Lockwood, C. J. et al. Preeclampsia-related inflammatory cytokines regulate

interleukin-6 expression in human decidual cells. Am. J. Pathol. 172, 1571–9 (2008). 61. Benichou, G., Gonzalez, B., Marino, J., Ayasoufi, K. & Valujskikh, A. Role of Memory

T Cells in Allograft Rejection and Tolerance. Front. Immunol. 8, 170 (2017). 62. Zeng, W. et al. Deep Surveying of the Transcriptional and Alternative Splicing

Signatures for Decidual CD8+ T Cells at the First Trimester of Human Healthy Pregnancy.

Front. Immunol. 9, 937 (2018).

63. Gehad, A. et al. A primary role for human central memory cells in tissue immunosurveillance. Blood Adv. 2, 292–298 (2018).

64. Zaph, C., Uzonna, J., Beverley, S. M. & Scott, P. Central memory T cells mediate long-term immunity to Leishmania major in the absence of persistent parasites. Nat. Med.

10, 1104–1110 (2004).

Referenties

GERELATEERDE DOCUMENTEN

expression of FOXP3 in first-trimester decidual tissue from pregnancies with a male fetus could imply an inferior maternal immune tolerance in early pregnancies with a male

We did not find differences in the levels of these cytokines between healthy pregnant and pree- clamptic women during pregnancy, suggesting that the reduced activated proportions

For this review, papers were included using combinations of the following search terms in Pubmed: ‘recurrent miscarriage, recurrent abortions, deciduas, immune, placenta,

Our findings do suggest that prednisolone either directly, or indirectly via dis- ruption of the maternal immune response, exerts programing effects on offspring phenotype, as shown

preeclamptic women in the decidua and lower expression on all memory T cell subsets in the peripheral blood of preeclamptic women compared to healthy pregnant women was shown..

Ook pro- porties van CD8 + geheugen T-cellen en CD8 + centrale geheugen T-cellen zijn lager in de decidua parietalis van vroege pre-eclampsie vergeleken met gezonde

Similar to mouse studies, in early pregnancy fetal maternal immune tolerance is probably not exclusively managed by memory Treg cells, since higher naive Treg cell subsets

Onderzoek naar geheugen T-cellen tijdens en na de zwangerschap is belangrijk, maar in de patiëntenpopulatie lijkt meer draagvlak voor onderzoek naar het geheugen tijdens en na