effect of parity on memory and regulatory t cells subsets...

40
Effect of parity on memory and regulatory T cells subsets in the decidua Scientific report Author: L.P.Zijlker [email protected] [email protected] S2485907 Supervisors: Dr. M.M. Faas 1 Dr. J.R. Prins 2 Department: 1 Department of Obstetrics and Gynecology 2 Department of Pathology and Medical Biology University Medical Center Groningen University of Groningen Research period: 21-09-2016 24-05-2017

Upload: others

Post on 22-Mar-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

Effect of parity on memory and regulatory

T cells subsets in the decidua Scientific report

Author:

L.P.Zijlker

[email protected]

[email protected]

S2485907

Supervisors:

Dr. M.M. Faas1

Dr. J.R. Prins2

Department: 1Department of Obstetrics and Gynecology

2Department of Pathology and Medical Biology

University Medical Center Groningen

University of Groningen

Research period:

21-09-2016 – 24-05-2017

Page 2: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

2

Page 3: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

3

Abstract

Introduction. During pregnancy, the maternal immune system acquires a unique state of

tolerance for the allogeneic fetus. The exact mechanisms responsible for this tolerance remain

unclear, however, modulation of the T-lymphocyte response at the fetal-maternal interface

appears to be important for a successful pregnancy. From previous research it can be

concluded that the maternal immune system can specifically recognise fetal antigens and

forms memory cells possibly specific for these fetal antigens. Furthermore, the risk of

developing immunological pregnancy complications is lower in subsequent pregnancies. In

this research we aimed to determine the influence of parity on memory and regulatory T cell

subsets in the human decidua.

Methods and Materials. Lymphocytes were enzymatically isolated from the decidua of

nulliparous (n=4), primiparous(n=5) and multiparous women (n=6). The lymphocytes were

stained with an antibody mix for characterisation of memory T cells and Tregs. Analysis was

done using flow cytometry.

Results. We found highly activated memory T cell subsets, but no significant differences

between the three groups . We found significantly higher proportions CD4+ and CD8+

activated Tregs in primiparous and multiparous women as well as suppressive CD4+ and

CD8+ Tregs. We also found significantly higher proportions of CD4+ and CD8+ memory

Tregs in primi- and multiparous women.

Conclusion. We conclude that parous women show a more activated and suppressive

regulatory T cell population at the fetal-maternal interface, as well as higher proportions

memory regulatory T cells. Further research is needed to investigate the effect of these

changes in regulatory T cell subsets on the development of immunological pregnancy

complications.

Page 4: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

4

Contents

Abstract ..................................................................................................................................... 3

Contents ..................................................................................................................................... 4

List of abbreviations ................................................................................................................. 6

1. Introduction .......................................................................................................................... 7

1.1 Immune associated complications of pregnancy .............................................................. 7

1.2 Immunology of pregnancy ................................................................................................ 7

1.2. Fetal-maternal interface ................................................................................................... 8

1.3 Adaptive immune system ................................................................................................. 8

1.3.1 Regulatory T-cells ...................................................................................................... 9

1.3.2 Memory T-cells .......................................................................................................... 9

1.4 Aim of this study. ........................................................................................................... 10

2. Methods and Materials ...................................................................................................... 11

2.1 Population ....................................................................................................................... 11

2.2 Anatomy of the placenta ................................................................................................. 11

2.3 Samples ........................................................................................................................... 11

2.4 Analysis .......................................................................................................................... 14

2.5 Gating strategy ................................................................................................................ 14

2.6 Statistics .......................................................................................................................... 16

3. Results ................................................................................................................................. 17

3.1 Demographics and clinical characteristics ..................................................................... 17

3.2 Memory T cells ............................................................................................................... 18

3.2.1 CD4+ memory cells ................................................................................................. 18

3.2.2 Activated CD4+ Memory T Cells ............................................................................ 19

3.2.3 CD8+ Memory T cells ............................................................................................. 20

3.2.4 Activated CD8+ memory T cells ............................................................................. 21

3.3 Tregs ............................................................................................................................... 22

3.3.1 CD4+ Treg cells ....................................................................................................... 22

3.3.2 CD4+ Memory Tregs ............................................................................................... 23

3.3.3 CD8+ Treg cells ....................................................................................................... 24

3.3.4 CD8+ memory Tregs ............................................................................................... 25

4. Discussion ............................................................................................................................ 26

4.1 Memory T cells ............................................................................................................... 26

Page 5: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

5

4.2 Tregs ............................................................................................................................... 27

4.2.1 Memory Tregs .......................................................................................................... 27

4.2.2 Suppressive Tregs .................................................................................................... 28

4.2.3 Activated Tregs ........................................................................................................ 28

4.3 Strengths and limitations ................................................................................................ 29

4.4 Conclusion ...................................................................................................................... 30

4.5 Future perspectives ......................................................................................................... 30

References ............................................................................................................................... 31

Acknowledgements ................................................................................................................. 35

Abstract Nederlands .............................................................................................................. 36

Appendix ................................................................................................................................. 37

Protocol Enzymatic isolation of lymphocytes from decidua tissue ...................................... 37

Page 6: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

6

List of abbreviations

APC Antigen presenting cell

B cell B-lymphocyte

BMI Body mass index

CM cell Central memory cell

CTLA-4 Cytotoxic T-lymphocyte-associated protein 4

DC Dendritic cell

EM cell Effector memory cell

FMO Fluorescence minus one

FOXP3 Forkhead box P3

FSC Forward scatter

HLA Human leukocyte antigen

IDO Indoleamine 2,3-dioxygenase

IQR Interquartile range

IUGR Intrauterine growth restriction

MHC Major histocompatibillity complex

MP Multiparous

NP Nulliparous

PBS Phosphate buffered saline

RM cell Tissue-resident memory cell

PP Primiparous

rpm Rounds per minute

RPMI 1640 Roswell Park Memorial Institute 1640 medium

SD Standard deviation

SSC Side scatter

RT Room temperature

T cell T-lymphocyte

TCR T cell receptor

Th T helper cell

Treg T regulatory cell

TRM Tissue-resident memory T cell

Page 7: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

7

1. Introduction

Pregnancy is a unique phenomenon, which presents an immunological challenge for the

maternal immune system. Under normal circumstances, the immune system rejects allogeneic

cells, such as transplants, tumours and microbes to protect the body from potential harm(1). A

fetus expresses HLA-antigens which are both maternal and paternal derived and the fetus is

therefore considered to be semi-allogeneic for the maternal immune system(2). However, the

maternal immune system does not reject the fetus but instead adapts and develops tolerance

towards it. Even while the maternal immune system tolerates the semi-allogeneic fetus, it can

still fight pathogens and infections, which illustrates the careful regulation of anti- and pro-

inflammatory responses by the immune system during pregnancy(2).

Though numerous studies have been conducted to identify the mechanisms responsible for the

development of tolerance of the maternal immune system towards the fetus, the exact

pathways are still unknown.

1.1 Immune associated complications of pregnancy

An inadequate adaptation of the maternal immune system during pregnancy is associated with

pregnancy complications such as preeclampsia, intrauterine growth restriction (IUGR) and

recurrent miscarriage(3–6). Preeclampsia is one of the leading causes of perinatal, maternal

and fetal morbidity and mortality, complicating 2-8% of pregnancies and contributing 16% of

maternal deaths in developed countries(4). The incidence of preeclampsia is higher in women

who are pregnant for the first time (primiparous women) compared to multiparous women

who have already experienced a normal pregnancy. However multiparous women have the

same risk of developing preeclampsia as a primiparous women when one of their subsequent

pregnancies is from a different father(3,7). Therefore it could be hypothesized that not the

state of pregnancy is memorized, but specifically paternal antigens.

This hypothesis is also supported by findings that prolonged exposure to seminal fluid, and

thus to paternal antigens, prior to the pregnancy is associated with a reduced risk at

preeclampsia and IUGR(8,9). How the memorizing competence of the maternal immune

system contributes to a healthy pregnancy has not been fully elucidated.

1.2 Immunology of pregnancy

The first hypothesis that tried to explain the unique phenomenon of tolerance for the

allogeneic fetus by Medawar (1953) was threefold: an absolute physical barrier between the

mother and the fetus, immunologically immature fetal antigens and inertness of the maternal

immune system prevent an attack on the fetus by the maternal immune system(10).

However this threefold hypothesis has been proven invalid over time. Although the fetal-

maternal barrier minimises immunological contact, it is not absolute. Fetal trophoblast cells

invade the uterine wall of the mother which then forms the maternal part of the placenta, the

decidua(11). Maternal immune cells which reside in the decidua therefore have direct access

to fetal antigens(12). Immunological contact between the mother and the fetus is also

demonstrated by research that found fetal antibodies to circulate in the blood of the mother,

even for years after pregnancy (microchimerism)(13). Maternal lymphocytes are able to

recognize fetal antigens through indirect presentation by antigen-presenting cells (APC)(14).

Therefore immaturity of fetal antigens is an insufficient explanation for the tolerance of the

Page 8: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

8

fetus. As for the inertness of the maternal immune system, studies have shown that activated

T cells specific for fetal antigens circulate in the peripheral blood of the mother(15–17)

1.2. Fetal-maternal interface

As concluded above, the maternal immune system has access to fetal antigens and can

actively recognize it. This suggests there are additional mechanisms which help to evade an

immune response of the maternal immune system against the fetus.

The fetus is connected to the mother by the placenta. In the placenta, nutrients, oxygen and

waste products are exchanged between the blood of the mother and the fetus(11). As

mentioned before, during gestation fetal trophoblast cells invade the uterine wall of the

mother. The multiple sites where fetal trophoblast cells are in direct contact with maternal

tissue are referred to as the fetal-maternal interface(12). At the fetal-maternal interface, the

maternal immune system is exposed to fetal antigens. The placenta plays an important role in

preventing rejection of the allogeneic fetus. Amongst others, cells of the innate immune

system are important in preventing an immune attack by the mother(12). The innate immune

system comprises the first, non-specific but quick response against microbes. Innate immune

cells such as macrophages, natural-killer cells (NK cells) and dendritic cells are abundant in

the placenta and essential for a successful pregnancy(18).

1.3 Adaptive immune system

As opposed to the innate immune system, the adaptive immune system is more specific but

has a slower response against foreign antigens. The adaptive immune system can be divided

into the humoral and the cellular response(1). The humoral response is mediated by B-

lymphocytes and focuses on extracellular immunity. The main mechanism of the humoral

response is the production of antibodies that can specifically attack circulating foreign

antigens, upon recognition by B-cells(1). The cellular immune response is mediated by T-

lymphocytes and comprises the intra-cellular immunity. T cells are able to recognize infected

cells and mediate in the destruction or actively destroy the infected cells themselves(1).

The T cell receptor (TCR) of T-lymphocytes recognizes antigens when presented on a major

histocompatibility complex (MHC). Within the T-lymphocyte population two subsets can be

distinguished, the CD4+ T-cells that are able to recognize antigens presented on a MHC-II

complex and CD8+ T cells that recognize antigens presented on a MHC-I complex. CD4+ T

cells, or T-helper cells release cytokines that regulate the immune response. CD8+ T cells, or

cytotoxic T cells are able to kill infected or damaged cells(1).

CD4+ T cells can be divided in different subsets, amongst others T-helper 1 (Th1), T-helper 2

(Th2) and T-helper 17 cells (Th17). Th1 cells stimulate a pro-inflammatory immune response

and are involved in auto-immune reactions while Th2 cells have an anti-inflammatory

function and are involved in allergic reactions. Th17 cells, like Th1 cells, mediate an

inflammatory response and are involved in auto-immunity. A shift in the balance between

Th1 and Th2 towards a Th2 dominance has been demonstrated in normal pregnancy(19). In

women with preeclampsia this Th1-Th2 shift was found absent, or even a shift towards Th1

was found(20). It was hypothesized that the Th1 down regulating and anti-inflammatory Th2

dominance was responsible for developing tolerance for the allogeneic fetus(21). However,

recent research found that this explanation does not fully cover the development of maternal

tolerance during the pregnancy. This was demonstrated amongst others by Th2 cytokine (Il-4

and Il-10) deficient mice which do not show a disturbed pregnancy(21–23).

Page 9: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

9

1.3.1 Regulatory T-cells

Regulatory T cells (Tregs) are specialised T cells that are potent immune-response

modulators. They are able to supress a cytotoxic T cells response, and are identified by the

expression of the transcription factor Foxp3(24). Tregs are involved in immunological self-

tolerance but there is also accumulating evidence that they play a key role in the

immunological acceptance of the allogeneic fetus(25–28). Tregs are able to suppress the Th1

and Th17 response, which has been noticed to be increased in women who suffered recurrent

miscarriage, and low percentages of Tregs were found in the blood of preeclamptic

women(27,29).

In healthy pregnancies, research has found high concentrations of CD4+ Tregs at the fetal-

maternal interface, that were able to suppress fetus-specific and fetus-nonspecific immune

responses(30). Furthermore, elevated levels of fetal-specific CD4+ Tregs that persist long

after pregnancy and a rapid expansion of Tregs during a subsequent pregnancy, indicate the

development of Treg memory cells(31).

1.3.2 Memory T-cells

Upon activation by an antigen, a small proportion of T cells differentiate into a memory cells.

These memory cells recognize the antigen at a second encounter and stimulate an even more

effective immune response. Different subtypes of memory T cells can be distinguished: the

effector-memory cells (EM), central-memory cells (CM) and tissue-resident memory cell

(TRM)(32)(Fig1). EM cells are memory cells that migrate to the site of inflammation and

show immediate effector function. CM cells reside in secondary lymph nodes and are not only

able to rapidly proliferate upon antigen stimulation, but also to renew themselves(33). Tissue-

resident memory cells are a special population of memory cells that remain permanently in

peripheral tissue even after infection(32).

Memory T cells express the cell surface marker CD45RO(34). Within the memory T cell

population CM cells can be identified by the chemokine receptor CCR7(32). When activated

they lose CCR7 and proliferate to EM cells, which express CD69(32). The TRM cells are

identified using the cell surface markers CD69 and CD103(Fig. 1)(32)(35).

Figure 1. Proliferation patterns of memory T cells(35)

Page 10: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

10

CD8+ EM cells are the most abundant type of T cells in the decidua(36,37). The

accumulation and activation of CD8+ EM cells found in the decidua during pregnancy, is

however not seen in peripheral blood(38). A previous study showed that in peripheral blood,

levels of CD4+ EM cells were higher during pregnancy compared to non-pregnant women.

The CD4+ EM, CM and activated memory T cells were elevated in parous women compared

to women who have never been pregnant (nulligravid women)(38). These findings support the

hypothesis that pregnancy is an immunological challenge which triggers the generation of

memory cells. The alteration of the memory T cell population could be an explanation for the

higher incidence of preeclampsia in primiparous women compared to multiparous women.

How one, or multiple pregnancies influence the memory T cell population at the fetal-

maternal interface, however is not fully known.

1.4 Aim of this study.

In this study, we aim to examine the effect of parity on the different subsets and

characteristics of memory T cells in human decidual tissue. The populations of memory T

cells in nulliparous, primiparous and multiparous women will be compared. Additionally, the

regulatory T cell populations will be determined and compared.

In previous research, elevated levels of CD4 central- and effector-memory cells have been

reported in peripheral blood after pregnancy(38). We therefore hypothesized that memory T

cell subsets are generated during first pregnancy and can rapidly expand during subsequent

pregnancy. Following these data we hypothesised to find higher levels of memory T cells with

tolerance competence in primiparous and multiparous women compared to nulliparous

women.

More knowledge about the memory T and Treg population at the fetal-maternal interface will

help to elucidate the mechanisms responsible for maternal immunological tolerance for the

allogeneic fetus. This knowledge could be implemented in the development of treatment or

prevention of immune associated adverse pregnancy outcome, preeclampsia, IUGR and

recurrent miscarriages.

Page 11: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

11

2. Methods and Materials

2.1 Population

The placentas of healthy women aged between 18 and 40, who were scheduled for an elective

caesarean section at >37 and < 42 weeks of gestation were included in this study. Women

with an immune deficiency, or those who used medication during their pregnancy were

excluded. All pregnancies were conceived in a natural way and if multiparous, all the

pregnancies were conceived by the same father according to the mothers. Four nulliparous,

five primiparous and six multiparous women were included. The women were classified

according to their parity at the time of inclusion. The included women that were expecting

their first child were classified as nulliparous, women expecting their second child were

classified as primiparous and all women pregnant for the third time or more, were classified

as multiparous. All women gave

informed consent and the study was

approved by the Medical Ethics

Committee of the UMC Groningen.

2.2 Anatomy of the placenta

The placenta consists of a fetal and a

maternal side. The maternal side,

called the decidua, can be divided in

the decidua parietalis and the decidua

basalis. The decidua parietalis is

maternal part which is connected to

chorioamniotic membrane and the

decidua basalis is the most outer part

of the basal plate which is connected

to fetal trophoblast cells (Fig. 2).

Isolation of lymphocytes from the

decidua parietalis and basalis enables

analysis of maternal lymphocytes that

were exposed to fetal antigens.

2.3 Samples

The placenta was collected

immediately after the caesarean-

section and brought to the lab.

Samples were taken from the maternal parts of the placenta.

The decidua basalis was obtained by cutting of the cotyledons of the basalis plate, which then

were washed in phosphate buffered saline (PBS) (PBS, without Mg and Ca, Lonza), and

cutting of the villi to get rid of the trophoblastic tissue. In order to obtain the decidua

parietalis, the amnion was removed and the chorioamniotic membrane was cut off the basal

plate and washed in PBS to rinse off blood clots. Small blood vessels were removed using

forceps. The decidua parietalis was scrapped of the chorion using a Costar® cellscraper

(Corning Incorporated).

Both tissues were minced and washed again. The tissue was then transferred into C-tubes

(Miltenyi Biotec) at 5ml tissue per tube and 10ml of StemProAccutase (Life Technologies)

was added.

Figure 2. Anatomy of the placenta

Page 12: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

12

The decidual tissue was mechanically dissected using

a Gentlemacs dissociator (Miltenyi Biotec) and

incubated in accutase in a water bath at 37°C for 45

minutes for enzymatic dissociation of the decidua

tissue. The enzymatically dissolved tissue was then

filtered through a 70 µm strainer with PBS. The

resulting cell suspensions were washed and 10 ml of

RPMI was added. For separation of the lymphocytes

from the other cells, a percoll gradient was used. The

gradient consisted of three layers at different

dilutions: 10 ml 40% (ρ=1.050 g/ml), 12,5 ml 45%

(ρ=1.056 g/ml) and 10 ml 68% (ρ=1.084 g/ml). To the

10 ml 40% dilution, 10 ml suspension was added and

layered on top of the 68% and 45% percoll dilution.

The percoll gradient was centrifuged (Thermo Fisher

Scientific, heraeus multifuge X3R) at 2000 rpm for 30

minutes without brake.

The lymphocyte layer between the 40% and 45%

dilution layers was taken off (Fig. 3) and the resulting

cell suspensions were washed with PBS. The number

of cells were counted using a coulter counter (Beckman).

Staining

The cells were transferred to a well plate at a million cells per well. First the cells were

blocked with 50 µl Fc-Block (BD Bioscience) and mouse serum (Sanquin, The Netherlands)

in PBS for 10 minutes at room temperature, to reduce aspecific binding of antibodies.

Subsequently the cells were stained with 50 µl (0.43 µg) FVS 620 (BD Biosciences) at room

temperature to distinguish between live and dead cells.

To identify Treg and memory T cell subsets, two different antibody mixes were used, both

containing 8 antibodies. 50 µl of each extracellular antibody mix (BD Biosciences) was added

to the cells and incubated for 30 minutes at 4°C (Table 1).

Figure 3. Percoll layers

Page 13: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

13

Table 1. Antibody panels (BD Biosciences)

A. Memory T cell antibody panel.

Antibody Concen

tration

Clone Fluorochro

me

Intra-/

extra-

cellular

Marking

CD4 1:100 SK3 APC-H7 Extra-

cellular

T-helper cells

CD8 1:200 RPA-T8 PE-Cy7 Extra-

cellular

Cytotoxic T-cells

CD45RO 1:20 UCHL-1 BV510 Extra-

cellular

Memory T-cells

CD103 1:50 BER-ACT8 BV421 Extra-

cellular

Tissue resident T-

memory cells

CCR7 1:10 150503 PE Extra-

cellular

Central memory T-

cells

CD69 1:20 FN50 APC Extra-

cellular

Activation maker

Foxp3 1:20 236A/E7 AF488 Intra-

cellular

Treg cells

FVS 620 1:50 PerCP-

Cy™5.5

Extra-

cellular

Viability

B. Regulatory T cell antibody panel.

Antibody Concen

tration

Clone Fluorochrome Intra-/

extra-

cellular

Marking

CD4 1:100 SK3 APC-H7 Extra-

cellular

T-helper cells

CD8 1:200 RPA-T8 PE-Cy7 Extra-

cellular

Cytotoxic T-cells

CD45RO 1:20 UCHL-1 BV510 Extra-

cellular

Memory T-cells

CD127 1:20 hIL-7R-

M21

PE Extra-

cellular

Negative for Treg

cells

HLA-DR 1:200 646.6 APC Extra-

cellular

Treg memory cell

CTLA-4 1:20 BNI3 BV421 Intra-

cellular

Suppressive function

marker

Foxp3 1:20 236A/E7 AF488 Intra-

cellular

Treg cells

FVS 620 1:50 PerCP-

Cy™5.5

Extra-

cellular

Viability

Page 14: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

14

Following extracellular staining, the cells were washed with PBS and fixed and permeabilised

with BD fix/perm for 40 minutes on ice. After permeabilisation the cells were washed twice

with BD Perm/wash buffer. The lymphocytes were then stained with an intracellular

antibody-mix for 30 minutes on ice before they were again washed twice with BD Perm/wash

buffer. The samples were diluted in 150 µl PBS before analysis (see appendices for full

protocol).

2.4 Analysis

The samples were analysed immediately after staining using a FACSVerseTM Flow

cytometer and BD FACS SuiteTM software (BD Biosciences). Compensation settings were

set using beads stained with a single antibody (UltraComp eBeads, eBioscience).

2.5 Gating strategy

To identify different sub-populations of T-cells the data were analysed using FlowJo v10

software. Gates were set using an unstained control, due to the low amount of isolated cells

gates could not be set using isotype controls or fluorescence minus one (FMO). However,

previous research in our group has shown that isotype controls did not vary much between the

experiments(38). First, a gate was set on the lymphocytes in a FSC/SSC scatterplot based on

size and granularity (Fig. 4A, Fig. 5B). In the lymphocyte population, the live lymphocytes

were selected in a histogram by setting a gate on the FVS negative cells (Fig. 4B, Fig. 5B).

The CD4+ and CD8+ T-cells were then displayed and gated in a scatterplot (Fig. 4C, Fig.

5C).

Memory T-cells

Within the CD4+ and CD8+ populations, the Foxp3- cells (Fig. 4D) and CD45RO+ cells (Fig.

4E) were selected in histograms to identify the memory T-cells. To identify different subsets

of T-memory cells, CD69 was used as an activated T-cell marker, CD103 as a tissue-resident

T-memory cell marker (Fig. 4F) and CCR7 was used as a central memory T-cell marker (Fig.

4G). This memory panel also permits identification of Tregs through selection of the Foxp3+

cells.

Regulatory T-cells

The regulatory T-cells can be identified by selecting the Foxp3+ and CD127- cells in the

CD4+ and CD8+ populations (Fig. 5D). CD45RO was used as a Treg memory cell marker,

CTLA-4 for suppressive antigen-experienced Tregs and HLA-DR as an activation marker

(Fig. 5E, Fig. 5F).

Page 15: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

15

a b c

d e f

g h i

Figure 4. Gating strategy to identify memory T-cells. FSC/SSC scatterplot of all events with gate set around

lymphocytes (A). The FVS negative cells are selected in a histogram to select the vital cells (B) In a scatterplot CD4+

and CD8+ are distinguished (C). The Foxp3 negative population is selected in a histogram (D). The CD45RO

population is selected in a histogram (E). In a scatterplot the CD69, CD103 and CCR7 populations can be distinguished

(F,G,H). The CCR7 and CD69 positive populations are shown in a scatterplot.

Page 16: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

16

2.6 Statistics

Data analysis was performed using IBM SPSS Version 23 data editor for Windows and

GraphPad Prism 5.04 software. Because of the small sample size, non-parametric distribution

of all data was assumed. Statistical differences between the three groups were tested using the

Kruskal-Wallis test where after the groups were compared pairwise using the Mann-Whitney

U test. Additionally the PP and MP group combined was compared to the NP group using a

Mann-Whitney U test. Differences between groups were considered statistically significant if

p<0.05. Because of the small groups, means instead of medians with interquartile range are

shown in the graphs.

Figure 5. Gating strategy to identify Tregs. FSC/SSC scatterplot of all events with gate set around lymphocytes (A).

The FVS negative cells are selected in a histogram to select the vital cells (B) In a scatterplot CD4+ and CD8+ are

distinguished (C). The Foxp3 positive population and CD127 negative population is selected in a histogram (D). In a

scatterplot the HLA-DR and CTLA-4 positive populations can be distinguished (E) and the CD45RO positive group is

selected in a scatterplot (F). In the CD45RO positive population, the HLA-DR and CTLA-4 population are shown (G)

a b c

d e f

g

Page 17: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

17

3. Results

3.1 Demographics and clinical characteristics

To examine the effects of parity on T cell subsets, 4 nulliparous, 5 primiparous and 6

multiparous women were included in this study. One decidua basalis sample in the

multiparous group was excluded because of incorrect handling in the lab. The patient

characteristics of these groups are shown in table 2.

There were no significant differences in maternal age or gestational age between the three

groups. The BMI of women in the nulliparous (NP) group was significantly lower than those

in the primiparous (PP) and multiparous (MP) group (p<0.05), however, there was no

significant difference between the PP and MP group. The birth weight of the babies in the NP

group was significantly lower compared to those in the PP group (p=0.05). No other

significant differences were found.

In all three groups there was one person who reported smoking during her pregnancy, none of

the women reported using alcohol during the pregnancy. All women received the same

antibiotics prior to the caesarean section. The pregnancies of multiparous women were

conceived by the same father as the previous pregnancies.

Table 2. Characteristics of the patient groups.

Nulliparous

(N=4)

Primiparous

(N=5)

Multiparous

(N=6)

Maternal age (years) 28 (22.8 ± 31.8) 30 (27 ± 34.5) 35 (27 ± 39)

BMI (kg/m2) 23.88 (20.6 ± 26.2)* 28.63 (27.1 ± 34.9) 32.98 (27.7 ±36.3)

Gestational age

(days)

275 (266 ± 277.3) 275 (270 ± 280) 274 (273 ± 275)

Parity 0 1 2.0 (2 ± 3)

Birth weight (gram) 3087.5

(2816.25 ± 3538.8)*

4500

(3787.5 ± 4765.5)

3607.5

(3306.3 ± 3701.3)

Fetal Sex

Boys

Girls

0.75

0.25

0.8

0.2

0.5

0.5 (Median ± interquartile range (IQR)). Fetal sex is shown as a percentage of the total number of inclusions. The

data was compared using a Kruskal-Wallis test, followed by a Mann-Whitney U test . *=p<0.05.

Page 18: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

18

3.2 Memory T cells

3.2.1 CD4+ memory cells

Percentages of CD4+ memory cells

were compared between nulliparous,

primiparous and multiparous women.

Figure 1 shows the proportion

CD4+CD45RO+ cells of CD4+ cells

found in the decidua parietalis and

decidua basalis. No significant

differences were found between the

groups.

CD4+ central memory cells

Central memory (CM) cells express

the lymph node homing receptor CCR7 (CD45RO+CCR7+)(32). There was no significant

difference in proportions CM cells between the three groups (Fig. 6A).

Figure 6. CD4+ memory cells in nulliparous, primiparous and multiparous women. CM cells, CD45RO+CCR7+

(A); EM cells, CD45RO+CCR7-(B); and RM cells, CD45RO+CD103+ (C) as a proportion of CD4+ cells. A

Kruskal-Wallis test was performed (p<0.05). Horizontal lines indicate means.

CD4+ CM cells

%C

D4

+C

D4

5R

O+

CC

R7

+/C

D4

+

bas par bas par bas par

0

10

20

30

40

50

NP PP MP

A CD4+ EM cells

%C

D4

+C

D4

5R

O+

CC

R7

-/C

D4

+

bas par bas par bas par

0

20

40

60

80

100

NP PP MP

B

CD4+ TRM cells

%C

D4

+C

D4

5R

O+

CD

10

3+

/CD

4+

bas par bas par bas par

0

5

10

15

20

NP PP MP

C

Figure 1. Proportions CD4+ memory T of CD4+ cells in decidua

basalis and parietalis of nulliparous, primiparous and multiparous

women. A Kruskal-Wallis test was performed (p<0.05). Horizontal

lines indicate means.

CD4+ memory cells

%C

D4

+C

D4

5R

O+

/CD

4+

bas par bas par bas par

0

20

40

60

80

100

NP PP MP

bas

par

Page 19: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

19

CD4+ EM cells

Effector memory cells lack the surface marker CCR7(32). There was no significant difference

in the proportions of effector memory cells with the phenotype CD4+CD45RO+CCR7-

between NP, PP and MP women (Fig. 6B).

CD4+ TRM cells

Resident memory cells are characterized by the cell surface marker CD103(32). There was no

significant difference in proportion CD4+CD45RO+CD103+ cells of CD4+ cells between

NP, PP and MP women (Fig. 6C).

3.2.2 Activated CD4+ Memory T Cells

Following activation, T cells express the cell surface marker CD69(35). Within the different

memory cell subsets, the proportions of activated cells were determined by comparing the

cells that stained positive for CD69 to the original population. However, no significant

differences were found between the groups for CD4+ memory cells, CM cells, EM cells or

RM cells (Fig. 7).

Figure 7. CD4+ activated memory cells in nulliparous, primiparous and multiparous women. Proportions

activated CD4+ memory cells of CD4+ memory cells(A); proportions activated CM cells of CM cells

(CD45RO+CCR7+CD69+) (B); proportions activated EM cells of EM cells (CD45RO+CCR7-CD69+)(C);

proportions activated RM cells of RM cells (CD45RO+CD103+CD69+)(D). A Kruskal-Wallis test was

performed (p<0.05). Horizontal lines indicate means.

Activated CD4+ memory cells

%C

D4

+C

D4

5R

O+

CD

69

+/C

D4

+C

D4

5R

O+

bas par bas par bas par

0

20

40

60

80

100

NP PP MP

A Activated CD4+ CM cells

%C

D4

+C

D4

5R

O+

CC

R7

+C

D6

9+

/

CD

45

RO

+C

CR

7+

bas par bas par bas par

0

20

40

60

80

100

NP PP MP

B

Activated CD4+ EM cells

%C

D4

+C

D4

5R

O+

CC

R7

-CD

69

+/

CD

45

RO

+C

CR

7-

bas par bas par bas par

0

10

20

30

40

50

NP PP MP

C Actvivated CD4+ TRM cells

%C

D4

+C

D4

5R

O+

CD

10

3+

CD

69

+/

CD

45

RO

+C

D1

03

+

bas par bas par bas par

0

20

40

60

80

100

NP PP MP

D

Page 20: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

20

3.2.3 CD8+ Memory T cells

CD8+ Memory cells

The CD8+ memory cell subsets were

determined using the same markers and

compared in the same way as CD4+ memory

cell subsets. No differences in percentage

CD8+ memory cells with a CD45RO+

phenotype were seen between the groups

(Fig. 8).

CD8+ CM cells

The percentage of CCR7+ CM cells

within the CD8+CD45RO+ population

was determined for NP, PP and MP

women. No significant differences were found between the groups (Fig. 9A).

Figure 9. CD8+ memory cells in nulliparous, primiparous and multiparous women. CM cells, CD45RO+CCR7+

(A); EM cells, CD45RO+CCR7-(B); and RM cells, CD45RO+CD103+ (C) as a proportion of CD4+ cells. A

Kruskal-Wallis test was performed, followed by a Mann-Whitney U test (#=p<0.1). Horizontal lines indicate

means.

CD8+ CM cells

%C

D8

+C

D4

5R

O+

CC

R7

+/C

D8

+

bas par bas par bas par

0

5

10

15

20

NP PP MP

A CD8+ EM cells

%C

D8

+C

D4

5R

O+

CC

R7

-/C

D8

+

bas par bas par bas par

0

20

40

60

80

100

NP PP MP

B#

CD8+ TRM cells

%C

D8

+C

D4

5R

O+

CD

10

3+

/CD

8+

bas par bas par bas par

0

20

40

60

NP PP MP

C

Figure 8. CD8+ memory T cells in nulliparous, primiparous and

multiparous women. A Kruskal-Wallis test was performed

(p<0.05). Horizontal lines indicate means.

CD8+ memory cells

%C

D8

+C

D4

5R

O+

/CD

8+

bas par bas par bas par

0

20

40

60

80

100

NP PP MP

Page 21: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

21

CD8+ EM cells

We found a trend of higher levels of effector memory cells with a CD8+CD45RO+CCR7-

phenotype cells in the decidua parietalis of NP women compared to PP women (p<0.1)(Fig.

9B).

CD8+ RM cells

There were no significant differences in CD8+CD45RO+CD103+ RM cell proportions of

CD8+ cells between NP, PP and MP women (Fig. 9C).

3.2.4 Activated CD8+ memory T cells

Activation in the CD8+ memory cell population activation was also examined using the

activation marker CD69. No significant differences were found between the groups for CD8+

memory cells, CM cells, EM cells or RM cells (Fig. 10).

Figure 10. CD8+ activated memory cells in nulliparous, primiparous and multiparous women. Proportions

activated CD8+ memory cells of CD8+ memory cells(A); proportions activated CM cells of CM cells

(CD45RO+CCR7+CD69+) (B); proportions activated EM cells of EM cells (CD45RO+CCR7-CD69+)(C);

proportions activated RM cells of RM cells (CD45RO+CD103+CD69+)(D). A Kruskal-Wallis test was

performed (p<0.05). Horizontal lines indicate means.

Activated CD8+ EM cells

%C

D8

+C

D4

5R

O+

CD

69

+/C

D4

5R

O+

bas par bas par bas par

0

20

40

60

80

100

NP PP MP

A Activated CD8+ CM cells

%C

D8

+C

D4

5R

O+

CC

R7

+C

D6

9+

/

CD

45

RO

+C

CR

7+

bas par bas par bas par

0

20

40

60

80

100

NP PP MP

B

CD8Temactivated

%C

D8

+C

D4

5R

O+

CC

R7

-CD

69

+/

CD

45

RO

+C

CR

7-

bas par bas par bas par

0

20

40

60

80

100

NP PP MP

C Activated CD8+ RM

%C

D8

+C

D4

5R

O+

CD

10

3+

CD

69

+/

CD

45

RO

+C

D1

03

+

bas par bas par bas par

0

20

40

60

80

100

NP PP MP

D

Page 22: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

22

3.3 Tregs

Regulatory T cells were identified by the combination of positive staining for the cell surface

marker Foxp3+ and negative staining for the cell surface marker CD127(35). The Treg cells

were only analysed in decidua basalis tissue because of the small amount of cells that could

be isolated from decidua parietalis tissue.

3.3.1 CD4+ Treg cells

The proportions of cells with a CD4+Foxp3+CD127- phenotype of CD4+ cells in decidua

basalis tissue were compared between NP, PP and MP women. No significant differences

were found between the groups (Fig. 11A).

Activated CD4+ Treg cells

Expression of HLA-DR indicates activation of the Treg cell(35). The proportion activated

Tregs of CD4+ cells was significantly lower (p<0.05) in the NP group compared to the PP

and MP group (Fig. 11B).

Suppressive CD4+ Treg cells

The immune-suppressive molecule CTLA-4 is expressed on antigen-experienced cells(35).

There were no significant differences in proportions CD4+Foxp3+CD127-CTLA-4+ cells of

CD4+ cells between the three groups (Fig. 11C).

Figure 11. CD4+ Tregs in decidua basalis tissue. Proportions Tregs (A), activated Tregs (B) and suppressive

Tregs (C) of CD4+ cells for NP, PP and MP women. A Kruskal-Wallis test was performed followed by a Mann-

Whitney U test (*=p<0.05). Horizontal lines indicate means.

CD4+ Treg cells

%C

D4

+F

oxp

3+

CD

12

7-/

CD

4+

np ppm

p

0

5

10

15

20

25

A Activated CD4+ Treg cells

%C

D4

+F

oxp

3+

CD

12

7-H

LA

-DR

+/C

D4

+

np ppm

p

0

20

40

60

80

100

B

*

Suppressive CD4+ Treg cells

%C

D4

+F

oxp

3+

CD

12

7-C

TL

A4

+/C

D4

+

np ppm

p

0

20

40

60

80

100

C

Page 23: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

23

3.3.2 CD4+ Memory Tregs

CD4+ memory Treg cells

Memory Treg cells were identified by their surface marker CD45RO(35). Between the three

groups there was no significant difference in percentage of CD4+Foxp3+CD127-CD45RO+

memory Tregs (Fig. 12A).

Activated CD4+ memory Treg cells

There was no difference in levels of memory Tregs with the phenotype CD4+Foxp3+CD127-

CD45RO+HLA-DR+ between the three groups. However, comparison of the NP to the MP

group using a Mann-Whitney U test, showed significantly lower levels of activated memory

Tregs in the NP group (p<0.05) (Fig. 12B).

Suppressive CD4+ memory Treg cells

No difference was found in levels of suppressive CD4+ memory Treg cells between the three

groups. However, when comparing the NP group to the PP and MP group combined, using a

Mann-Whitney U test, the proportion suppressive CD4+ memory Treg cells of CD4+ cells in

the NP group was significantly lower (p<0.05) (Fig. 12C).

Figure 12. CD4+ memory Tregs in decidua parietalis tissue. Proportions memory Tregs of CD4+ cells (A),

proportions activated memory Tregs of memory Tregs (B) and proportions suppressive memory Tregs of

memory Tregs (C) for NP, PP and MP women. A Kruskal-Wallis test was performed followed by a Mann-

Whitney U ((*)= p<0.05 for comparison of NP to PP/MP). Horizontal lines indicate means.

CD4+ memory Treg cells

%C

D4

+F

oxp

3+

CD

12

7-C

D4

5R

O+

/CD

4+

np ppm

p

50

60

70

80

90

100

A Activated CD4+ memory Treg cells

%C

D4

+F

oxp

3+

CD

12

7-C

D4

5R

O+

HL

A-D

R+

/

CD

4+

Fo

xp

3+

CD

12

7-C

D4

5R

O+

np ppm

p

0

10

20

30

40

B

( )*

Suppressive CD4+ memory Treg cells

%C

D4

+F

oxp

3+

CD

12

7-C

D4

5R

O+

CT

LA

4+

/

CD

4+

Fo

xp

3+

CD

12

7-C

D4

5R

O+

np ppm

p

0

20

40

60

80

100

C

( )*

Page 24: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

24

3.3.3 CD8+ Treg cells

Proportions cells with a CD8+Foxp3+CD127- phenotype of CD8+ cells in decidua parietalis

tissue were compared between NP, PP and MP women. No significant differences were found

between the groups (Fig. 13A).

Activated CD8+ Treg cells

There was no significant difference in proportions of CD8+ activated Treg cells between the

three groups (Fig. 13B).

Suppressive CD8+ Treg cells

We found a trend of higher levels of suppressive CD8+ Tregs with a CD8+Foxp3+CD127-

CTLA-4+ phenotype in MP women compared to NP women (p<0.1)(Fig. 13C). When

comparing only the NP group to the MP group using a Mann-Whitney U test, the proportion

of suppressive CD8+ Treg cells was significantly lower in the NP group (p<0.05).

Figure 13. CD8+ Tregs in decidua parietalis tissue. Proportions Tregs (A), activated Tregs (B) and suppressive

Tregs (C) of CD8+ cells for NP, PP and MP women. A Kruskal-Wallis test was performed followed by a Mann-

Whitney U ((*)= p<0.05 for comparison of NP to PP/MP). Horizontal lines indicate means.

CD8+ Treg cells

%C

D8

+F

oxp

3+

CD

12

7-/

CD

8+

np ppm

p

0

5

10

15

20

A Activated CD8+ Treg%

CD

8+

Fo

xp

3+

CD

12

7-H

LA

-DR

+/C

D8

+

np ppm

p

0

10

20

30

40

50

B

Suppressive CD8+ Treg cells

%C

D8

+F

oxp

3+

CD

12

7-C

TL

A4

+/C

D8

+

np ppm

p

0

20

40

60

80

100

C

( )*

Page 25: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

25

3.3.4 CD8+ memory Tregs

There were no significant differences in levels of CD4+Foxp3+CD127-CD45RO+ memory

Tregs between the three groups (Fig. 14A).

Activated CD8+ memory Treg cells

There were no significant differences in levels of activated memory Tregs with the phenotype

CD8+Foxp3+CD127-CD45RO+HLA-DR+ between the three groups (Fig. 14B).

Suppressive CD8+ memory Treg cells

No differences were found in levels of suppressive CD8+ memory Treg cells between the

three groups. However, when comparing only the NP to the MP group using a Mann-Whitney

U test, levels of suppressive memory Tregs were significantly lower in the NP group

(p<0.05)(Fig. 14C).

Figure 14. CD8+ memory Tregs in decidua parietalis tissue. Proportions memory Tregs of CD8+ cells (A),

proportions activated memory Tregs of memory Tregs (B) and proportions suppressive memory Tregs of

memory Tregs(C) for NP, PP and MP women. A Kruskal-Wallis test was performed followed by a Mann-

Whitney U ((*)= p<0.05 for comparison of NP to PP/MP). Horizontal lines indicate means.

CD8+ memory Treg cells

%C

D8

+F

oxp

3+

CD

12

7-C

D4

5R

O+

/CD

8+

np ppm

p

0

20

40

60

80

A Activated CD8+ memory Treg cells%

CD

8+

Fo

xp

3+

CD

12

7-C

D4

5R

O+

HL

A-D

R/

CD

8+

Fo

xp

3+

CD

12

7-C

D4

5R

O+

np ppm

p

0

10

20

30

40

50

B

Suppressive CD8+ memory Treg cells

%C

D8

+F

oxp

3+

CD

12

7-C

D4

5R

O+

CT

LA

4+

/

CD

8+

Fo

xp

3+

CD

12

7+

CD

45

RO

+

np ppm

p

0

20

40

60

80

100

C

*

Page 26: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

26

4. Discussion

In this study we examined the effect of parity on the subsets and characteristics of memory T

cells in decidual tissue. The effect of parity on memory T cell subsets was tested by

comparing lymphocytes extracted from decidual tissue of nulliparous, primiparous and

multiparous women. Additionally we determined and compared regulatory T cell subsets.

Our results show no differences in memory cell subsets between the three groups.

Interestingly parity does seem to have an effect on the Treg cell population. We found

significantly higher proportions of CD4+ and CD8+ Tregs with an activated and suppressive

phenotype as well as memory Tregs in primiparous and multiparous women compared to

nulliparous women.

4.1 Memory T cells

Contrary to what we hypothesized, we did not find higher proportions memory T cells or

memory T cells subsets in primiparous and multiparous women compared to nulliparous

women. There could be several reasons why we did not find higher levels of memory cells in

multiparous women:

Previous research showed that during pregnancy, maternal CD8+ T-lymphocytes can be

primed by fetal antigens and both lymphocytes and fetal antigens persist in the maternal blood

post-partum(15,38,39). A study of Barton et al. studied these antigen-experienced cells in a

mouse model performing re-exposure to fetal antigens by means of a subsequent

pregnancy(40). They observed no enhanced expansion of fetal-primed CD8+ memory cells

during pregnancy in parous mice. Our findings appear to be in line with these data, as we also

did not find higher levels of CD8+ memory T cells in the decidua of parous women. Barton et

al. found that pregnancy in mice induces T cells with a dysfunctional effector phenotype that

exhibit impaired cytokine production(40). During a subsequent pregnancy, these T cells

exhibited limited proliferative capacity. The limited proliferative capacities of the pregnancy

induced memory T cells could be an explanation for the lack of higher proportions of those

memory cells in multiparous women. More research examining proliferative capacities of

human memory T cells isolated from pregnant women in cell proliferation assays, could

provide very useful information.

A study by Kieffer et al. found significantly higher proportions of CD4+ and CD8+ effector-

memory T cells, CD4+ central-memory cells and activated CD4+ memory T cells in the

peripheral blood of non-pregnant multiparous women compared to nulligravid women. Our

results do not show increased levels of memory T cell subsets in the decidua of primiparous

or multiparous women. There could be several reasons why parity seems to increase the levels

of memory T cells in the peripheral blood, and not in the decidua:

the placenta features several specific characteristics which constrain a T cell mediated

response against fetal antigens. At the fetal-maternal interface, where fetal trophoblast cells

are directly exposed to the maternal immune system, large scale antigen recognition by

maternal T cells is not possible because trophoblast cells do not express classic MHC

molecules which are essential for direct antigen-recognition(1,41). Only indirect antigen

recognition mediated by maternal antigen-presenting cells enables maternal T cells to detect

the alloantigens(14,40).The expression of IDO by trophoblast cells also constrains an attack

of the maternal immune system because IDO inhibits rapid proliferation of T cells(23,42).

Trophoblast cells also express Fas ligand, which induces apoptosis in Fas-expressing

Page 27: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

27

activated CD4+ and CD8+ T lymphocytes(23,43). Finally, complement activation which

promotes a pro-inflammatory immune response is inhibited by expression of the Crry-protein

on trophoblast cells(23,44).

All together these mechanisms minimise a T cell mediated immune response against the

allogeneic fetus at the fetal-maternal interface. Perhaps these mechanisms also prevent an

influx of memory T cells that reside in the peripheral blood, into the decidua during a second

pregnancy(45). This would explain why higher proportions memory T cells are seen in the

peripheral blood of multiparous women but do not correspond with higher proportions

memory T cells in the decidua of multiparous women.

Another explanation for the relatively equal levels of memory cells in the decidua of

nulliparous, primiparous and multiparous women could be that memory cells migrate near the

end of pregnancy. During pregnancy, levels of T cells are increased in the decidua compared

to the peripheral blood, but post-partum increased levels of memory T cells are found in the

peripheral blood(36,38). Possibly, memory T cell migrate from the decidua towards the

peripheral blood at the end of pregnancy where they reside until antigen re-exposure during

the next pregnancy.

Alternatively, memory T cells subsets could diminish in the third trimester under influence of

hormonal changes. Progesterone is up regulated in the third trimester and has been shown to

suppress the T cell response, especially CD4+ effector activity(46).

Both of these explanations suggest that analysis of term decidua tissue will not show

differences in memory T cell subsets. Perhaps a difference can be found in decidua tissue in

the first or second trimester, however this is obviously difficult in healthy human pregnancies.

To our knowledge this is the first study to compare memory T cells in decidua tissue between

nulliparous, primiparous and multiparous women. Therefore we cannot compare our findings

to other studies.

4.2 Tregs

We found higher levels of activated CD4+ Tregs in multiparous and primiparous women, and

higher levels of CD8+CTLA-4+ memory Treg cells in multiparous women compared to

nulliparous women. When comparing only the nulliparous to the multiparous group

additionally higher levels of activated CD4+ memory Tregs, CD4+CTLA-4+ memory Tregs

and CD8+CTLA-4+ Tregs were found in multiparous women.

Regulatory T cells are potent suppressive immune regulators and are extensively studied for

their role in pregnancy. Previous research described an accumulation of CD4+ Tregs in the

decidua as well as antigen-independent systemic accumulation during pregnancy(25,30)

4.2.1 Memory Tregs

Memory T cells are antigen experienced cells that rapidly proliferate upon re-exposure to an

antigen and thereby protect against harmful pathogens during a subsequent exposure.

Regulatory T cells are also able to memorise antigens, these antigen experienced Tregs can

suppress a T cell response against a specific antigen after re-exposure(31,35). Memory Tregs

are probably very useful in establishing a rapid tolerance for the fetus in a subsequent

pregnancy(28,31).

Page 28: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

28

In this study, we found higher levels of memory Tregs with an activated phenotype in

primiparous and multiparous women compared to nulliparous women (Fig. 12B, Fig. 12C,

Fig. 14C). These data could support the hypothesis that memory Tregs are important in

establishing immunological tolerance in a subsequent pregnancy.

Our findings are in line with a mouse study by Rowe et al. that showed fetal-antigen specific

Tregs persisted at higher levels post-partum and rapidly re-expanded during a subsequent

pregnancy in the peripheral blood of mice(31). Interestingly these mice had a lower risk of

pregnancy complications in their second pregnancy compared to mice in which Tregs were

inhibited. As we previously mentioned, in humans the risk of pre-eclampsia is higher in a first

pregnancy. Furthermore, lower percentages of Tregs are seen in pre-eclamptic women(29).

Our data support the importance of Tregs in healthy pregnancies and show that multiparous

women exhibit higher percentages of Tregs in the decidua. Taken all together, perhaps

memory Tregs could be involved in the reason why the risk of pre-eclampsia is lower in

secondary pregnancies. Comparing the memory Treg population in the decidua of healthy

women to pre-eclamptic women could provide more insight into the role of memory Tregs in

pre-eclampsia.

4.2.2 Suppressive Tregs

We found an increased percentages of CD4+ and CD8+ Tregs as well as CD4+ and CD8+

memory Tregs that express CTLA-4 in multiparous women compared to primiparous women

(Fig. 12C, 13C, 14C). CTLA-4 is a marker of suppressive function which is up regulated in

Tregs after activation by antigens(47).

The significantly higher levels of Tregs expressing CTLA-4 we found in multiparous women,

indicate a subset of Tregs with higher functional capacities. This could explain why we did

not find higher levels of memory T cells in multiparous women as we hypothesized. CTLA-

4+ Tregs can attenuate antigen-specific T cell responses by competitive binding of co-

stimulatory molecules which are necessary for antigen-specific activation of T cells(48,49).

Also CTLA-4 has been shown to induce IDO expression on APC’s which, as we mentioned

previously, inhibits rapid T cell proliferation(50–52)

Therefore we propose that in multiparous women, the abundance of CTLA-4+ Tregs could

suppress the expansion of memory T cells that were formed during a previous pregnancy.

However, more research investigating the functional capacities of decidual Tregs is needed.

The importance of CTLA-4+ Tregs in fetal-maternal immune tolerance can be illustrated by

research that showed that CTLA-4 is down regulated on decidual Tregs in miscarriage

compared to normal pregnancy and in spontaneous abortion compared to induced

abortion(53).

4.2.3 Activated Tregs

The cell surface marker HLA-DR is a late activation marker. However, HLA-DR+ Tregs were

also identified as a distinct Treg subset with a high suppressive function(54). Our results show

higher levels of activated CD4+ Tregs and CD4+ memory Tregs in primiparous and

multiparous women compared to nulliparous women(Fig. 11B, Fig. 12B). A decrease of

HLA-DR+ expression on Tregs was seen in the peripheral blood of women in preterm

labour(55). Perhaps these high levels of HLA-DR+ Tregs are involved in mechanisms that

protect multiparous women against immunological pregnancy complications. However as

Page 29: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

29

mentioned above, more research investigating the functional capacities of decidual Tregs is

needed.

There is no definitive consent on phenotypical characterization of Tregs. In this study Foxp3

and low CD127 expression were used to define Tregs. CD25Bright

is also a commonly used

marker for CD4+ Treg cells in decidual tissue. CD4+CD25+ T cell subsets show regulatory

capacities, however CD25 is also expressed on effector T cells(13,15,16). CD25 therefore is

not an exclusive Treg cell marker. Foxp3+ was found to be expressed on cells with regulatory

function within the CD25+ as well as the CD25- T cell subsets(15,17,18). Foxp3 is now

considered to be a more specific marker for regulatory T cells.

As shown in mouse models, CD127 is not down regulated in all Tregs, which would make

CD127- an unreliable marker for Treg characterization. However, we have used CD127- as an

additional marker next to Foxp3. The combination of these two markers has been shown to

specifically identify the Treg cell population(56).

Tregs in decidual tissue have been intensively studied for their role in immune tolerance for

the fetus, the majority of this research has used CD25+ for phenotypic characterization of

Tregs. This marker was not included in our research as we believe the use of Foxp3 is more

accurate. This should be kept in mind when comparing our findings to earlier research on

Tregs.

4.3 Strengths and limitations

Strengthening this study, we analysed memory T cell subsets at the site of the immunological

challenge, the fetal-maternal interface. The cells present in the decidua are directly involved

in the immune response towards the fetus and therefore analysis of these subsets show us

which cells play an important role in the regulation of the immune response towards the fetus.

Furthermore, we included the placentas of women who gave birth by an elective caesarean

section. During labour the release of hormones, such as progesterone, and contractions

influence lymphocyte subsets(46). The delivery of the baby by a caesarean section minimises

the influence of labour on the lymphocytes. Also the protocol of isolation of the lymphocytes

from the placenta was started immediately after delivery, which minimised the time for

lymphocytes to change or deteriorate once oxygen supply is cut off.

Another strength of this study are the extensive antibody panels that were used to determine

the memory T cell and Treg cell subsets. Within the memory cell panel, the central-memory

cell population, effector-memory cell population and resident memory cell population could

be determined as well as the proportions of activated cells within these population. The Treg

cell panel included several markers for Tregs and additional suppression and activation

markers.

Several limitations should also be considered. First of all, due to the small number of elective

caesarean sections in the study period, only a small number of placenta’s were available for

this study. Therefore these results cannot be assumed to be representative for the general

population, however they give an indication of what could be expected if more patients were

included in this research. Also because the groups are relatively small, the data show a high

dispersion. Statistical analysis can therefore only show large differences. If more patients

were to be included, statistical significant differences between the groups could be examined

with greater reliability and precision.

Also, because of the small number of patients available for this research, we did not exclude

patients based on their BMI even though BMI is known to influence the immune

Page 30: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

30

response(57). However, only the BMI of the women in the nulliparous group was statistically

significantly different from the other groups, no statistical significant difference was found

between the primiparous and multiparous group.

Furthermore we analysed lymphocytes that were extracted from term decidual tissue. The

lymphocyte populations present in decidual tissue are known to change dynamically during

pregnancy. This study only allows conclusions about the proportions of different cell subsets

present at the end of pregnancy, but these might be different in earlier stages of pregnancy.

Isolation of lymphocytes from decidual tissue is more complicated than from peripheral

blood, and the amount of cells obtained is much lower. This limits possibilities for analysis of

these cells and prohibited us from assessing the functional features of these cells.

4.4 Conclusion

In conclusion, the results of this study show that parity did not affect memory T cell subsets in

the decidua as we did not find differences in memory T cell levels between the groups.

However, parity did seem to affect regulatory T cell subsets in the decidua. We found higher

levels of CD4+ and CD8+ Treg and memory Treg cells in primiparous and multiparous

women compared to nulliparous women. Furthermore, we found higher levels of antigen

experienced Tregs with a suppressive phenotype in primiparous and multiparous women

compared to nulliparous women.

This study contributes to existing knowledge on the development of immunological tolerance

for the fetus by showing that parity affects regulatory T cells population at the fetal-maternal

interface. Whether these alterations in the regulatory T cells population at the fetal-maternal

interface are involved in lowering the risk of immune associated complications of pregnancy

remains to be investigated.

4.5 Future perspectives

To assess the role of memory T cells and Tregs in maternal-fetal immune tolerance, more

research is required. First, more patients should be included in this study for more powerful

and reliable statistical analysis. Also studying the migration patterns of memory T cells

circulating in the peripheral blood could provide useful insight in the role of memory T cell in

maternal fetal immune tolerance. Next to the migration patterns of memory cells, the

proliferative and functional capacities of regulatory T cells isolated from human decidua

tissue could provide more insight into the role of regulatory T cells in immunological

tolerance for the allogeneic fetus.

Page 31: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

31

References

1. T.G. Rijkers, F.G.M. Kroese, C.G.M. Kallenberg RHWMD. Immunologie. 1e druk.

Bohn Stafleu van Loghum; 2012.

2. Robertson, SA., Petroff, MG., Hunt J. Knobil and Neill’s Physiology of reproduction.

4th editio. Elsevier; 2015. 1835-74 p.

3. Dekker G. The partner ’ s role in the etiology of preeclampsia. J Reprod Immunol.

2002;57:203–15.

4. Steegers EAP, Von Dadelszen P, Duvekot JJ, Pijnenborg R. Pre-eclampsia. Lancet.

2010;376(9741):631–44.

5. Guerin LR, Prins JR, Robertson SA. Regulatory T-cells and immune tolerance in

pregnancy: A new target for infertility treatment? Hum Reprod Update.

2009;15(5):517–35.

6. Southcombe JH, Mounce G, McGee K, Elghajiji A, Brosens J, Quenby S, et al. An

altered endometrial CD8 tissue resident memory T cell population in recurrent

miscarriage. Sci Rep. 2017;7(41335).

7. Robillard PY, Hulsey TC, Périanin J, Janky E, Miri EH, Papiernik E. Association of

pregnancy-induced hypertension with duration of sexual cohabitation before

conception. Lancet. 1994;344(8928):973–5.

8. Saftlas AF, Rubenstein L, Prater K, Harland KK, Field E, Triche EW. Cumulative

exposure to paternal seminal fluid prior to conception and subsequent risk of

preeclampsia. J Reprod Immunol. 2014;101–102(1):104–10.

9. Robertson SA, Sharkey DJ. Seminal fluid and fertility in women. Fertil Steril.

2016;106(3):511–9.

10. Medawar PB. Some immunological and endocrinological problems raised by the

evolution of viviparity in vertebrates. Soc Exp Biol. 1953;7:320–338.

11. Moffett A, Loke C. Immunology of placentation in eutherian mammals. Nat Rev

Immunol. 2006;6(8):584–94.

12. PrabhuDas M, Bonney E, Caron K, Dey S, Erlebacher A, Fazleabas A, et al. Immune

mechanisms at the maternal-fetal interface: perspectives and challenges. Nat Immunol.

2002;16(4):237–43.

13. Bianchi DW, Zickwolf GK, Weil GJ, Sylvester S, DeMaria M a. Male fetal progenitor

cells persist in maternal blood for as long as 27 years postpartum. Proc Natl Acad Sci U

S A. 1996;93(2):705–8.

14. Erlebacher A, Vencato D, Price KA, Zhang D, Glimcher LH. Constraints in antigen

presentation severely restrict T cell recognition of the allogeneic fetus. J Clin Invest.

2007;117(5):1399–411.

15. Van Kampen CA, Versteeg-van Der Voort Maarschalk MFJ, Langerak-Langerak J,

Van Beelen E, Roelen DL, Claas FHJ. Pregnancy can induce long-persisting primed

CTLs specific for inherited paternal HLA antigens. Hum Immunol. 2001;62(3):201–7.

Page 32: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

32

16. Tafuri A, Alferink J, Möller P, Hämmerling GJ, Tafuri A, Alferink J, et al. T Cell

Awareness of Paternal Alloantigens During Pregnancy. Science. 1995;270(5236):630–

3.

17. Jiang S, Vacchio MS. Cutting Edge: Multiple Mechanisms of Peripheral T Cell

Tolerance to the Fetal Allograft’’. J Immunol. 1998;160(7):3086–90.

18. Triggianese P, Perricone C, Chimenti MS, De Carolis C, Perricone R. Innate Immune

System at the Maternal-Fetal Interface: Mechanisms of Disease and Targets of Therapy

in Pregnancy Syndromes. Am J Reprod Immunol. 2016;1–13.

19. Raghupathy R. Pregnancy: success and failure within the Th1/Th2/Th3 paradigm.

Semin Immunol. 2001;13(4):219–27.

20. Saito S, Sakai M. Th1/Th2 balance in preeclampsia. J Reprod Immunol.

2003;59(2):161–73.

21. Chaouat G. The Th1/Th2 paradigm: Still important in pregnancy? Semin

Immunopathol. 2007;29(2):95–113.

22. White, C. A., Johansson M et al. Effect of Interleukin-10 Null Mutation on Maternal

Immune Response and Reproductive Outcome in Mice. Biol Reprod. 2003;70(1):123–

31.

23. La Rocca C, Carbone F, Longobardi S, Matarese G. The immunology of pregnancy:

Regulatory T cells control maternal immune tolerance toward the fetus. Immunol Lett.

2014;162(1):41–8.

24. Wang YM, Alexander SI. CD8 regulatory T cells: What’s old is now new. Immunol

Cell Biol. 2009;87(3):192–3.

25. Aluvihare VR, Kallikourdis M, Betz AG. Regulatory T cells mediate maternal

tolerance to the fetus. Nat Immunol. 2004;5(3):266–71.

26. Zenclussen AC, Schumacher A, Zenclussen ML, Wafula P, Volk H-D. Immunology of

pregnancy: cellular mechanisms allowing fetal survival within the maternal uterus.

Expert Rev Mol Med. 2007;9(10):1–14.

27. MacLean MA, Shilito J, Walker JJ, D M. Evidence of a T H 1 type response associated

with recurrent miscarriage. Fertil Steril. 2000;73(6):1206–8.

28. Clark DA. The importance of being a regulatory T cell in pregnancy. J Reprod

Immunol. 2016;116:60–9.

29. Prins JR, Boelens HM, Heimweg J, Heide S Van Der, Dubois AE, Oosterhout AJ Van,

et al. Preeclampsia is Associated with Lower Percentages of Regulatory T Cells in

Maternal Blood. Hypertens pregnancy. 2009;28:300–11.

30. Tilburgs T, Roelen DL, van der Mast BJ, de Groot-Swings GM, Kleijburg C, Scherjon

SA, 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. 2008;180(8):5737–45.

31. Rowe JH, Ertelt JM, Xin L, Way SS. Pregnancy imprints regulatory memory that

Page 33: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

33

sustains anergy to fetal antigen. Nature. 2012;490(7418):102–6.

32. Mueller SN, Gebhardt T, Carbone FR, Heath WR. Memory T Cell Subsets, Migration

Patterns, and Tissue Residence. Annu Rev Immunol. 2013;31:137–61.

33. Sallusto F, Geginat J, Lanzavecchia A. Central Memory and Effector Memory T Cell

Subsets: Function, Generation, and Maintenance. Annu Rev Immunol.

2004;22(1):745–63.

34. Sallusto F, Lenig D, Förster R, Lipp M, Lanzavecchia a. Two subsets of memory T

lymphocytes with distinct homing potentials and effector functions. Nature.

1999;401(6754):708–12.

35. Rosenblum MD, Way SS, Abbas AK. Regulatory T cell memory. Nature.

2015;16(2):90–101.

36. Tilburgs T, Schonkeren D, Eikmans M, Nagtzaam NM, Datema G, Swings GM, et al.

Human decidual tissue contains differentiated CD8+ effector-memory T cells with

unique properties. J Immunol. 2010;185(7):4470–7.

37. Tilburgs T, Strominger JL. CD8+ Effector T Cells at the Fetal-Maternal Interface,

Balancing Fetal Tolerance and Antiviral Immunity. Am J Reprod Immunol.

2013;69(4):395–407.

38. Kieffer TEC, Faas MM, Scherjon SA, Prins JR. Pregnancy persistently affects memory

T cell populations. J Reprod Immunol. 2017;119:1–8.

39. Lissauer D, Piper K, Goodyear O, Mark D, Moss PAH, Moss PAH. Fetal-Specific

CD8+ Cytotoxic T Cell Responses Develop during Normal Human Pregnancy and

Exhibit Broad Functional Capacity. J Immunol. 2017;189:1072–80.

40. Barton BM, Xu R, Wherry EJ, Porrett PM. Pregnancy promotes tolerance to future

offspring by programming selective dysfunction in long-lived maternal T cells. J

Leukoc Biol. 2017;101(4):975–87.

41. Arvelund JS. Immune regulation at the fetal ‐ maternal interface with focus on decidual

macrophages. In 2015.

42. Munn DH, Zhou M, Attwood JT, Bondarev I, Conway SJ, Marshall B, et al. Prevention

of allogeneic fetal rejection by tryptophan catabolism. Science. 1998;281(5380):1191–

3.

43. Hunt JS, Vassmer D, Ferguson TA, Miller L. Fas ligand is positioned in mouse uterus

and placenta to prevent trafficking of activated leukocytes between the mother and the

conceptus. J Immunol. 1997;158(9):4122–8.

44. Xu C, Mao D, Holers VM, Palanca B, Cheng a M, Molina H. A critical role for murine

complement regulator crry in fetomaternal tolerance. Science. 2000;287(5452):498–

501.

45. Patrice, N., Tagliani, E., Tay, C., Asp, p., Levy., DE., Erlebacher. A. Chemokine gene

silencing in decidual stromal cells limits T cell access to the maternal-fetal interface.

Science (80- ). 2012;336(6086):1317–21.

Page 34: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

34

46. Hughes GC, Clark E a, Wong AH. The intracellular progesterone receptor regulates

CD4+ T cells and T cell-dependent antibody responses. J Leukoc Biol.

2013;93(3):369–75.

47. Rosenblum MD, Gratz IK, Paw JS, Lee K, Marshak-Rothstein A, Abbas AK. Response

to self antigen imprints regulatory memory in tissues. Nature. 2011;480(7378):538–42.

48. Chambers CA, Kuhns MS, Egen JG, Allison JP. CTLA-4 Mediated Inhibition in

Regulation of T cell Responses: Mechanisms and Manipulation in Tumor

Immunotherapy. Annu Rev Immunol. 2001;19:565–94.

49. Oderup C, Cederbom L, Makowska A, Cilio CM, Ivars F. Cytotoxic T lymphocyte

antigen-4-dependent down-modulation of costimulatory molecules on dendritic cells in

CD4+ CD25+ regulatory T-cell-mediated suppression. Immunology. 2006;118(2):240–

9.

50. Grohmann U, Orabona C, Fallarino F, Vacca C, Calcinaro F, Falorni A, et al. CTLA-4-

Ig regulates tryptophan catabolism in vivo. Nat Immunol. 2002;3(11):1097–101.

51. Heikkinen J, Möttönen M, Alanen A, Lassila O. Phenotypic characterization of

regulatory T cells in the human decidua. Clin Exp Immunol. 2004;136(2):373–8.

52. Munn, DH., Sharma, MD., Lee, JR., Jhaver, KG., Johnson, TS., Keskin, DB., Marshall,

B., Chandler, P., Antonia, SJ., Burgess, R., Slingluff Jr. CL., Mellor A. Potential

Regulatory Function of Human Dendritic Cells Expressing Indoleamine. Science.

2002;297(5588):1867–70.

53. Jin LP, Chen QY, Zhang T, Guo PF, Li DJ. The CD4+CD25bright regulatory T cells

and CTLA-4 expression in peripheral and decidual lymphocytes are down-regulated in

human miscarriage. Clin Immunol. 2009;133(3):402–10.

54. Baecher-Allan C, Wolf E, Hafler DA. MHC class II expression identifies functionally

distinct human regulatory T cells. J Immunol. 2006;176(8):4622–31.

55. Kisielewicz A, Schaier M, Schmitt E, Hug F, Haensch GM, Meuer S, et al. A distinct

subset of HLA-DR+-regulatory T cells is involved in the induction of preterm labor

during pregnancy and in the induction of organ rejection after transplantation. Clin

Immunol. 2010;137(2):209–20.

56. Santegoets SJAM, Dijkgraaf EM, Battaglia A, Beckhove P, Britten CM, Gallimore A,

et al. Monitoring regulatory T cells in clinical samples: consensus on an essential

marker set and gating strategy for regulatory T cell analysis by flow cytometry. Cancer

Immunol Immunother. 2015;64:1271–86.

57. Ilavská S, Horváthová M, Szabová M, Nemessányi T, Jahnová E, Tulinská J, et al.

Association between the human immune response and body mass index. Hum

Immunol. 2012;73(5):480–5.

Page 35: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

35

Acknowledgements

First of all, I would like to thank my supervisors dr. Marijke Faas and dr. Jelmer Prins for

making this project possible. Especially I would like to thank dr. Prins and Tom Kieffer for

their help and guidance, and for always being available to answer our questions.

For all the long hours in the lab together, for your positive and patient attitude and for the fun

we have had, I would like to thank Anne Laskewitz. I would not have wanted to do this

project without you.

Also I would like to thank Bart de Haan for his guidance in the laboratory as well as the

immunology and endocrinology research group.

Finally, I would like to thank all the patients included in this research for their participation.

Page 36: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

36

Abstract Nederlands

Introductie. Tijdens de zwangerschap bestaat er een unieke situatie waarbij het moederlijk

immuunsysteem de semi-allogene foetus niet aanvalt. Hoe deze tolerantie ontstaat is nog niet

volledig opgehelderd, maar een aangepaste reactie van T-lymfocyten speelt zeker een rol. Uit

onderzoek blijkt dat er tijdens de zwangerschap T-geheugen cellen specifiek voor het foetale

antigeen worden aangemaakt. Ook is bekend dat een aantal immunologische

zwangerschapscomplicaties vaker voorkomen bij de eerste zwangerschap. Dit onderzoek

heeft tot doel de invloed van pariteit op de verschillende T-geheugen cel en de regulatoire T-

cel populaties in de placenta te bepalen.

Materiaal en methode. Lymfocyten zijn enzymatisch geïsoleerd uit de decidua van nulliparae

(n=4), primiparae (n=5) en multiparae (n=6). De lymfocyten zijn gekleurd met antilichamen

en geanalyseerd door middel van flow cytometrie.

Resultaten. We vonden geen verschil in de geheugen T-cel populaties tussen de drie groepen.

Significant hogere proporties geactiveerde en suppressieve regulatoire T-cellen waren te zien

in primiparae en multiparae. Daarnaast vonden we ook significant hogere proporties

regulatoire T-geheugencellen in primi- en multiparae.

Conclusie. In conclusie hebben we geen effect van pariteit op geheugen T-cel populaties

gevonden. Het doormaken van meerdere zwangerschappen lijkt te zorgen voor een meer

geactiveerde en suppressieve regulatoire T-cel populatie in de decidua en daarnaast te zorgen

voor hogere percentages regulatoire T-geheugen cellen. Verder onderzoek is nodig om te

bepalen wat het effect van deze veranderingen is op het ontwikkelen van immunologische

zwangerschapscomplicaties.

Page 37: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

37

Appendix

Protocol Enzymatic isolation of lymphocytes from decidua tissue

Date:

Preparation prior to experiment: Reserve laminar airflow cabinet

Bring Accutase to room temperature

PBS

RPMI

Antibodies

Fc block + mouse serum

Fix/Perm and Perm wash buffer

Basket and PBS to collect the placenta

Percoll solution

40% → 13.2 ml SIP + 19.8 ml RPMI

45% → 18.6 ml SIP + 22.7 ml PBS

68% → 22.4 ml SIP + 10,6 ml RPM

Isolation protocol

Preheat waterbath, collect: scissors, forceps, measuring cup, petri dish, filter, cell- scraper,

pipette, 50ml tubes, gloves, PBS, accutase, filters and C-tubes. Turn on radio.

Cut maternal side of decidua basalis, avoid damaged (white, yellow) tissue.

Cut the superfluous villi of the decidua basalis, keep the tissue in immersed in PBS while

preparing.

Remove the amniotic membrane from the chorion and cut the chorion from the basal plate.

Use the cellscraper to separate the decidua parietalis from the chorion, keep the tissue

immersed in PBS while preparing.

Take a sample of basalis and parietalis tissue and tranfser to 0.5ml RNAlater for parietalis and

1 ml RNAlater for basalis.

Transfer tissue to 50ml tubes and add PBS. Mince the tissue using scissors.

Centrifugeer 5 minutes at 1000 rpm. Remove supernatant.

Transfer tissue to C-tubes at 5 ml per tube and resuspend in 10 ml accutase at room

temperature.

Put the C-tubes in the GentleMacs dissociator.

For basalis choose: m-heart 02.01 (17 sec 668 rpr)

For parietalis choose: h-tumor 02 2x (37 sec 235 rpr)

Put the C-tubes in the waterbath and incubate for 45

minutes at 37°C while gently shaking.

After incubation, add 10 ml PBS to ach tube.

Filtrate over a 70 µm Smart Strainer in a 50 ml tubes.

Centrifuge 5 minutes at 1500 rpm at room temperature

Remove supernatant and resuspend in 10 ml RPMI.

Add 10 ml of 40% Percoll solution and resuspend.

Add 10 ml of 68% Percoll solution to a new 50 ml tubes. Slowly layer 12.5 ml of the 45%

solution on top of the 68% Percoll solution. Keep the tube at a 45º angle while layering. Then

layer 20 ml of the 40% Percoll / cell solution on top of the 45% layer in the same way.

Centrifuge for 30 minutes at 2000 rpm without brake.

Take of layer A, then collect the macrophage layer using a 1 ml pipette and transfer to a 50

ml tube. Next, collect the lymphocyte layer using a 1 ml pipette and transfer to a 50 ml tube.

Page 38: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

38

Add up PBS to 50 ml and centrifuge for 5 minutes at 1500 rpm at room temperature.

Resuspend in 1 ml of PBS

Count cells using the coulter counter.

Protocol Flow cytometry

Wells needed for experiment:

14 wells

6 lymphocytes

4 macrophages

4 single stains

Preparations before staining

Make 500 µl 1% Fc block + 10% mouse serum:

50µl per well

Add 445µl Facs buffer + 5µl Fcblock + 50µl mouse serum

Make 1200µl BD Fix/Perm working solution 1:4 (keep cold)

200 µl per well

300µl Fix/perm + 900µl diluent

Make 4,8 ml Perm/Wash buffer 1:5 (keep cold)

800µl per well

960µl of Perm/Wash buffer + 3840µl demi

Make FVS

10 µl FVS + 490 µl PBS

Cell preparation Add 1.000.000 cells per well in 96 well plates according to scheme, 3.000.000 cells for

macrophages

Spin down at 1800 RPM for 3min (RT) and discard supernatant

Stain with 50ul FVS for 15 min at RT in the dark

Wash twice with PBS (2min, 1800, RT).

Block cells with 50 µl 1% FC block + 10 % mouse serum for 10 minutes (RT)

Spin down at 1800 RPM for 3 min (4C) and discard supernatant

Add 50µl extracellular antibodymix to all wells according to scheme

Incubate on ice in the dark for 30 min.

Spin down at 1800 RPM for 3 min (4C) and discard supernatant

Wash 2X with 200µl PBS

Fixating cells

Fix all samples in 200 µl BD fix/perm in the fridge for 40 min.

Spin down at 1800 RPM for 3 min(4C) and discard supernatant

Wash 2x with 200 µl Perm wash buffer

Resuspend macrophages in 150 µl PBS and keep dark in the fridge until analysis

Only for Tregs & Tmem

Add 50 µl of intracellular antibodymix to all wells according to scheme

Incubate on ice in the dark for 30min

Wash 2x with 200 µl Perm wash buffer

Resuspend the samples in 150 µl PBS

Keep cells in the dark in the fridge until analysis

Page 39: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

39

Staining protocol

1

2

3

4

5

6

7 8 9 10 11 12

A par Tregs Tmem Cntrl ss FVS

B

C Macro Cntrl ss FVS

D

E bas Tregs Tmem Cntrl ss FVS

F

G Macro Cntrl ss FVS

H

Cell count

Basalis lymfo Basalis macro

1ml 1ml

1E6 1E6

Parietalis lymfo Parietalis macro

1ml 1ml

1E6 1E6

Antibody Panels Extracellulair

Tregs

AB Intra/extra Verdunning Volume (x2) Kanaal

CD4 Extra 1:100 1 µl APC-H7

CD8 Extra 1:200 0.5 µl Pe-Cy7

CD45RO Extra 1:20 5 µl V500-C

CD127 Extra 1:20 5 µl PE

HLA-DR Extra 1:200 0.5 µl APC

PBS 88 µl

Tmem

AB Intra/extra Verdunning Volume

(2x)

kanaal

CD4 Extra 1:100 1 µl APC-H7

CD8 Extra 1:200 0.5 µl Pe-Cy7

CD45RO Extra 1:20 5 µl V500-C

CD103 Extra 1:50 2 µl BV421

CCR7 Extra 1:10 10 µl PE

CD69 Extra 1:20 5 µl APC

PBS 76.5 µl

Page 40: Effect of parity on memory and regulatory T cells subsets ...scripties.umcg.eldoc.ub.rug.nl/FILES/root/geneeskunde/2017/zijlkerlp/... · pregnancy complications such as preeclampsia,

40

Macrophages

AB Extra Verdunning Volume

(2x)

Kanaal

HLA-DR Extra 1:10 10 µl APC

CD163 Extra 1:50 2 µl BV421

CD14 Extra 1:20 5 µl V500-C

CD209 Extra 1:20 5 µl FITC

ICAM3 Extra 1:50 2 µl PE

CD80 Extra 1:20 5 µl Pe-Cy7

CD45 Extra 1:20 5 µl APC-H7

PBS 66 µl

Intracellulair

Treg

AB Intra/extra Verdunning Volume (x2) Kanaal

CTLA-4 Intra 1:20 5 µl BV421

FOXP3 Intra 1:20 5 µl AF488

PBS 90 µl

Tmem

AB Intra/extra Verdunning Volume

(2x)

kanaal

FOXP3 Intra 1:20 5 µl AF488

PBS 95 µl

Beads

Vortex beads (30 seconds).

Divide 1 drop over 1 tubes (30 µl per tube ).

Add 0.5 µl antibody.

Incubate for 30 minutes in the fridge.

Wash 2 times with 200 µl PBS (5min, 1800 rpm) and remove supernatant using a pipette tip.

Resuspend in 150 µl PBS, keep cold in the dark until analysis.