Sie sind auf Seite 1von 19

CHAPTER  8

Immunology of Normal Pregnancy


and Preeclampsia

CHRISTOPHER W.G. REDMAN, IAN L. SARGENT AND ROBERT N. TAYLOR

Editors’ comment: Chesley in his single-authored first edi- subsequent pregnancies. There is also the issue of partner
tion wrote little regarding immunology and preeclampsia, specificity and primipaternity.2,3
though he noted that as early as 1902 Viet had suggested
that deported trophoblastic fragments are antigenic and
could elicit antibodies that he named ‘syncytiolysin (edi- MATERNAL ADAPTATION
tion 1 p. 467). He further noted that Dienst had suggested TO A FOREIGN FETUS
incompatibility of maternal and fetal blood groups in 1905
(edition 1 p. 470). Despite such early beginnings the first Maternal immune accommodation to paternal antigens
edition’s index had a little over 30 citations under the sub- expressed by their fetus might be acquired from a previ-
heading “Immunologic Factors.” Perhaps this represented ous successful pregnancy or abortion, in the case of the
immunology’s state of the art at that time or, as the cur- same partner, or exposure to paternal seminal plasma.
rent chapter’s authors imply, investigators then aware that Partner specificity (primipaternity) was first suggested
preeclampsia rarely reoccurred in subsequent conceptions when a change of partner by parous women seemed to
were dissuaded from exploring immunological factors increase the risks of preeclampsia.2 This study and oth-
in the genesis of preeclampsia. However, our knowledge ers were anecdotal or uncontrolled but suggested that the
of the intricacy and complexity of the immune system has issue was relevant. Subsequent reports seemed to con-
expanded since Chesley’s first edition, as the authors of firm this (for example ref. 3) but a systematic review con-
this chapter show us. This report was, to our knowledge, cluded that there are still substantial uncertainties.4 This
the first to suggest a role for placental debris, or microves- was confounded by the finding that longer inter-pregnancy
icles, in preeclampsia. This initially made many an inves- intervals are associated with both a change of partner and
tigator smile, but microvesicles are now being appreciated preeclampsia5,6 Which was the relevant variable: a new
everywhere, and are elegantly discussed here, as are other partner or delayed conception? One possible modifier is
aspects of growing information regarding the immunologi- cigarette smoking, which is known to be negatively asso-
cal significance of preeclampsia. ciated with preeclampsia but positively associated with a
change of partner.5 Statistical adjustment for these asso-
ciated factors has not been necessarily helpful because
INTRODUCTION of confusion about the causal relationships of associated
variables.7
In this chapter we describe the role of immune mecha- The concept that maternal immune adaptation to a
nisms in preeclampsia. The issue has always centered partner’s fetus might not be restricted to pregnancy but be
on the need to explain why women in their first pregnan- learnt before conception by exposures to sperm or seminal
cies are most susceptible to the condition, which has been fluid (reviewed by Saito et  al. 20078) was stimulated by a
combined with the perception that the presence within the study from Guadeloupe, which showed that a short interval
mother of a genetically foreign fetus must pose a challenge between first coitus and conception significantly increased
to the maternal immune system.1 It has been postulated that the risk of preeclampsia9 regardless of parity but specifi-
immune accommodation to the fetus needs to be “learnt” cally with a new partner (Table 8.1). A more recent inves-
or immunoregulated. In preeclampsia the adaptation may tigation12 confirms the issue of the short interval but only
be relatively defective in a first pregnancy but less so in primiparous women were studied.

Chesley’s Hypertensive Disorders in Pregnancy. DOI: http://dx.doi.org/10.1016/B978-0-12-407866-6.00008-0


ISBN: 978-0-12-407866-6 161 © 2015
2014 Elsevier Inc. All rights reserved.
162 Chesley’s Hypertensive Disorders in Pregnancy

TABLE 8.1  Prepregnancy Priming of Maternal TABLE 8.2  Danger Signals that Activate the Innate
Immune System (Inflammatory) Immune System
Preeclampsia risk Stimulants of Inflammation Corresponding Receptors

First pregnancy Short duration of ↑1 Bacterial products Toll-like receptors


coitus, pre-conception Products of oxidative stress Scavenger receptors
Previous term New partner ↑2 Products of cell trauma Toll-like and scavenger
pregnancy Thrombin receptors
Previous abortion Short previous Probably↓3 Heat shock proteins Protease-activated receptors
pregnancy Soluble (viral) DNA Various
Barrier Reduced exposure to Possibly↑4 Toll-like receptor 9
contraception sperm/seminal fluid:
Intracytoplasmic No prior immune Possibly↑5
sperm injection exposure to sperm/ or viruses. When inflammatory cells are activated they
seminal fluid release signals such as cytokines or chemokines that, in
Donor insemination No prior exposure to Probably↑6 turn, attract and “instruct” adaptive immune cells (T or B
sperm/seminal fluid lymphocytes) to generate antigen-specific responses by
Donor oocyte Fully allogeneic fetus Probably↑6 means of antibodies or cytotoxic cells. Hence the two sys-
Sources:10,11 tems, innate and adaptive, operate together and in sequence.
1
Compared to first pregnancy with longer exposure. Adaptive immunity develops slowly but delivers precise
2
Compared to second pregnancy with same partner. antigen-specific responses with immunological “memory.”
3
Compared to no previous abortion. It evolved more recently than innate immunity and is pres-
4
Barrier contraception compared to no barrier contraception.
5
Compared to no prior exposure to sperm/seminal fluid.
ent only in vertebrates. The innate and adaptive systems
6
Compared to matched control group. are asymmetrically interdependent; in effect the latter is a
functional elaboration of the former. The innate system
does not need the adaptive system to function whereas
In summary there is indirect evidence that tolerization adaptive immunity has an absolute requirement for signals
to a foreign fetus can occur in prior pregnancies or even from the innate system. Adaptive immunity is strongly but
before conception, but if tolerance is not achieved, pre- not exclusively influenced by the so-called “transplanta-
eclampsia may ensue. Such tolerization would be expected tion antigens,” the human leukocyte antigens (HLA), which
to involve the adaptive immune system. However the more determine transplant rejection. The distribution of fetal (for-
primitive innate immune system, which controls inflamma- eign) HLA on trophoblast is therefore a key issue that has
tory responses, is also involved. informed much of our insights into pregnancy immunol-
ogy (see below). The main effectors of adaptive immunity
are T cells and B cells. They are triggered by professional
INNATE AND ADAPTIVE IMMUNITY antigen-presenting cells expressing class II MHC antigens
(HLA-D) that enable them to activate naïve T cells. They
Innate and adaptive immunity both contribute to the patho- comprise dendritic cells, macrophages, and B cells.
genesis of preeclampsia. Innate immunity is a more primi- In this chapter the inflammatory system is deemed to
tive, rapid-acting, early-response system for global and be the sum of events involving innate rather than adaptive
relatively nonspecific protection. It evolved to protect single immune changes. It is one component of a much larger
and multicellular organisms from “danger.”13 Danger takes system that responds to all cellular stresses, called the inte-
many forms: physical, for example high temperature, chem- grated stress response.15
ical, for example excessive oxidation, trauma, infection, or
neoplasia. Of these, oxidative stress is a relevant proinflam-
matory trigger in both normal pregnancy and preeclampsia. NATURE’S TRANSPLANT
The innate immune system depends on a network of
“danger” receptors, called pattern recognition receptors Within the fetoplacental unit, the placenta is the tissue that
(PRRs), which are germ-line encoded and recognize many is most directly exposed to maternal immune cells, which
different danger signals. Their ligands may be endogenous, have contact with trophoblast in the placental bed, inter-
often in the form of modified self-molecules, or exogenous, villous space and amniochorion. Fetal cells may traffic
typically derived from bacteria or viruses (Table 8.2). The across the placenta but in general are confined to the fetal
same receptor frequently recognizes multiple antigens.14 compartment.
Most danger receptors are either soluble or bound to the Maternal adaptive immunity appears to be most impor-
cell surface but some function intracellularly, for example tant during placentation, when the placenta and its uteropla-
binding intracellular ligands that are derived from bacteria cental circulation become established during the first stage
Chapter 8 · Immunology of Normal Pregnancy and Preeclampsia 163

Classical Non-Classical
Class Ia Class Ia

Tissue HLA-A HLA-B HLA-C HLA-E HLA-G

Adult and fetal cells

Extravillous,cytotrophoblast
(Interface 1)

Syncytiotrophoblast
(Interface 2)

FIGURE 8.1  The unusual distribution of Class 1 HLA antigens on trophoblast. Human trophoblast does not express Class II (HLA-D)
antigens. Syncytiotrophoblast expresses no HLA.

of preeclampsia. The major contribution to the maternal Interface 2 is anatomically distinct and comprises syn-
syndrome in the second stage of preeclampsia appears to cytiotrophoblast, the surface layer of chorionic villi, which
derive from a systemic (or vascular) inflammatory response is in contact with maternal blood borne immune cells. The
(innate immunity). interface becomes active when the intervillous circulation is
A key aspect of the maternal–placental immune inter- established (weeks 8–10) and expands with the growth of
actions is the restricted expression of HLA by tropho- the placenta to become the dominant interface towards the
blast. Trophoblast has a uniquely different pattern of HLA end of pregnancy. Because the syncytium is in direct con-
expression from other somatic cells. The decidual (extravil- tact with the maternal circulation, if it promoted immune
lous) trophoblast in the placental bed does not express the responses they would be systemic, not local. At Interface 2,
strong polymorphic HLA-A, HLA-B (HLA-Class I) or the syncytiotrophoblast is HLA-negative and immunologi-
HLA-D (HLA-Class II) antigens, the principal stimulators cally neutral.
of T-cell-dependent graft-rejection responses. Instead, it The key point is that paternal HLA alloantigens are only
expresses a unique combination of HLA-C and non-classi- expressed at Interface 1, which is most active in the first
cal HLA-type 1b antigens: HLA-E and HLA-G (Fig. 8.1). half of pregnancy. This constrains the space and time when
Of these only HLA-C is polymorphic, meaning that it alone abnormal placentation can cause the later development of
expresses paternal identity. HLA-E is a ubiquitous sig- preeclampsia and fetal growth restriction.
nal for self, which inhibits NK cells via specific receptors.
Recognition of “missing-self” is immunostimulatory and
contributes to immune recognition of malignant cells lack- CLASSICAL TWO-STAGE MODEL
ing HLA-E.16 HLA-G is not expressed in normal tissues OF PREECLAMPSIA
other than the placenta, where it is confined to extravillous
trophoblast.17 It is also expressed in some malignancies and In 1991 we first proposed that preeclampsia evolves in two
may contribute to escape from immune surveillance. It is stages, pre-clinical and clinical21 (Fig. 8.3). Each stage
generally agreed that it confers a degree of immune privi- involves the maternal immune system in different ways. In
lege on trophoblast.18 A soluble form of HLA-G is also the first stage there is an important element localized to the
released and is detectable in blood. placental bed where placentation is typically inhibited. In
Maternal exposure to trophoblast varies with gestational the second stage, a diffuse vascular inflammation predomi-
age. The maternal-fetal immune interfaces19 are not fully nates. The two stages are associated with different patterns
characterized but include two that are relevant to preeclamp- of maternal exposure to fetal tissues and potentially differ-
sia, which we have called Interfaces 1 and 2 (Fig. 8.2), and ent consequences of immune maladaptation.
one that might be relevant but is largely unstudied, namely Immune Interface 1 needs to be considered in relation to
the chorionic component of the placental membranes. Stage 1 preeclampsia and immune events that affect placen-
Interface 1 is between maternal immune cells and inva- tation could explain its apparent partner specificity. Immune
sive, extravillous trophoblast in the decidua of the placental Interface 2 is active through trimesters 2 and 3 and seems to
bed. It dominates during the first half of pregnancy when be the dominant component of Stage 2 preeclampsia.
placentation is established (see Chapter 5) and the placental The two stage model has now been superseded by a more
bed is infiltrated with invasive cytotrophoblast. detailed multi-staged model as explained in a later section.
164 Chesley’s Hypertensive Disorders in Pregnancy

Decidua

Villi and intervillous space

Interface 1 Interface 2

Mononuclear Multinuclear
cytotrophoblast syncytiotrophoblast
Extravillous
trophoblast

FIGURE 8.2  The two immune interfaces of human pregnancy. Adapted from20 with permission. Immune events at Interface 1 drive
the first stage of the two-stage model (see Fig. 8.3) of preeclampsia, whereas Interface 2 drives the second stage. Interface 1 comprises
maternal immune cells, including uterine NK cells (uNK), T lymphocytes (L), macrophages (M) and dendritic cells (DC), and the invasive
extravillous cytotrophoblast (Tx), between large stromal cells (S) and spiral arteries (SPA; not to scale). Interface 2 lies between circulating
maternal immune cells, including T lymphocytes (L), NK cells (NK), monocytes (Mo) and dendritic cells (DC), and syncytiotrophoblast
microparticles (STBM). (This figure is reproduced in color in the color plate section.)

Two stages of preeclampsia and


two maternal-fetal immune interfaces
Stage 1 Maternal
Poor placentation
First half of pregnancy interface 1

Oxidatively stressed
placenta

sFlt-1 and other mediators


Stage 2 Maternal
Second half of
pregnancy interface 2
Dysfunctional maternal
endothelium

Clinical signs of preeclampsia


FIGURE 8.3  Two-stage model of development of preeclampsia. (This figure is reproduced in color in the color plate section.)
Chapter 8 · Immunology of Normal Pregnancy and Preeclampsia 165

STAGE 1 PREECLAMPSIA, INTERFACE 1 with KIRs more strongly than HLA-C126 so the combina-
AND MATERNAL IMMUNE RESPONSES tion of maternal HLA-C2 with fetal KIR B/B could be the
TO TROPHOBLAST best for promoting adequate placentation and avoiding
preeclampsia. This is what is observed. Kir AA mothers
The endometrium is an immune tissue. During the luteal confronted with HLA-C2 fetuses are the most susceptible
phase it differentiates and begins to transform by infiltra- to preeclampsia,27,28 a similar pattern being shown with
tion of leukocytes. Once early pregnancy is established, recurrent spontaneous miscarriages and normotensive fetal
decidua leukocytes are mainly (75%) natural killer cells. growth restriction.29
Macrophages comprise a smaller, but still abundant popula- This form of maternal-fetal immune recognition can
tion. There are also rare dendritic cells and T cells. B cells explain the partner specificity for preeclampsia but not the
are conspicuous by their absence. Natural killer cells are protection conferred by a previous pregnancy by the same
part of the innate immune system. Uterine NK (uNK) cells partner, namely the immunological memory. As far as is
differ in phenotype from most circulating NK cells, are known, immune memory is provided only by the adaptive
more activated, less cytotoxic and have an enhanced capac- immune system.
ity to secrete cytokines and angiogenic factors. The latter The issue with respect to trophoblast is one of T-cell
promote infiltration of the spiral arteries by invasive tro- recognition of HLA-C. HLA-C has unique features that
phoblast.22 uNK cells bear receptors that interact with the distinguish if from HLA-A and B. It is less polymorphic
unique repertoire of HLA expressed by invasive cytotropho- for example, and in general its surface expression is lower.
blast in the placental bed (HLA-C, -E and -G). HLA-C, the However, trophoblast uniquely proves the exception to this
only polymorphic HLA expressed by trophoblast, appar- rule. Foreign antigens are recognized by T cells when they
ently confined to invasive cytotrophoblast, is the ligand for are bound to HLA-A and -B proteins on the cell surface of
killer immunoglobulin-like receptors (KIR) expressed by antigen-presenting cells – so-called HLA-restriction. HLA-
uNK cells. The main receptors for HLA-G are the inhibi- C-restricted presentation of antigens has been less readily
tory leukocyte immunoglobulin-like receptors (LIR -1 and demonstrable except in specific viral infections and a few
LIR-2) which are expressed by monocytes, NK cells, T self-peptides. It is important that in the decidua, fetal (pater-
cells, and macrophages.23 It is crucial to the issue of part- nal) HLA-C, expressed by trophoblast, can bind to both NK
ner specificity of preeclampsia that HLA-C can signal fetal and T cells (although not coincidentally). It is also relevant
paternity, which can be recognized by the uNK cells. that the KIR receptors that dominate perceptions of inter-
The KIR receptors that recognize HLA-C on invasive actions between uNK cells and invasive cytotrophoblast are
trophoblast are themselves extremely polymorphic such that also expressed by T cells where they appear to be able to
two individuals are unlikely to have the same genotype. The modulate (inhibit or enhance) T cell responses.30
variability is not simply that of polymorphic genes, but of Maternal T cell responsivenes to foreign fetal HLA-C
different patterns of inheritance of up to 17 different genes has been detected in the deciduas of normal pregnancies
each with its own polymorphism, some that activate, others but is thought to be kept in check by T-regulatory cells.31
that inhibit. The number of KIR genes in different geno- Although this is a key issue, T cell reactivity to HLA-C on
types varies. The expression of the genotype is itself vari- invasive cytotrophoblast in preeclampsia remains undefined.
able through differential expression of KIR genes, which Techniques in assisted reproduction create new immune
becomes fixed by methylation, with the phenotype passed to challenges for pregnant women, with hitherto novel immune
daughter cells.24 HLA-C has more than 1000 haplotypes.25 mating combinations from donated gametes, sperm, oocytes
In other words, maternal-fetal immune recognition at the or embryos. Some methods of assisted reproduction
site of placentation involves two polymorphic gene systems, increase the risk of preeclampsia. Such modes of concep-
maternal KIRs and fetal HLA-C molecules. Hence partner tion or a short interval between first coitus and conception
specificity will be high, if not unique (see Chapter 4). may hinder T-cell-dependent tolerance to paternal antigens,
KIR haplotypes can be divided into two groups, A and which facilitates the establishment of normal pregnancy at
B, the latter being distinguished by additional activating Interface 1. Otherwise, abnormal placentation and uteropla-
(presumably beneficial) receptors. It is presumed that uNK cental perfusion can lead to abnormalities at Interface 2 and
cells need to be activated to produce cytokines and angio- Stage 2 disease.
genic factors to promote placentation. A maternal haplotype
B would be predicted to protect from preeclampsia. In any
pregnancy, the maternal KIR genotype could be AA (no STAGE 2 PREECLAMPSIA AND INTERFACE 2
activating KIR) or AB/BB (presence of one or more activat-
ing KIRs). Normal pregnancy and preeclampsia are both associated
HLA-C haplotypes can also be grouped as C1 and C2 with a low-grade systemic (vascular) inflammatory response,
depending on an amino acid dimorphism at one position in which is more intense in preeclampsia.32 It is believed this
the alpha-1 domain. It is considered that HLA-C2 interacts is secondary to syncytiotrophoblast stress33 induced by
166 Chesley’s Hypertensive Disorders in Pregnancy

hypoxia, oxidative stress or both. A generalized response to Protein folding consumes energy. Not surprisingly ER
any form of cellular or tissue damage is inflammation,13 a stress is precipitated by energy deficiency, for example from
hypothesis which has been subsequently validated in many hypoxia or glucose deprivation. The ISR has profound effects
different contexts, not associated with pregnancy. In pre- on metabolism that help cells survive under stress and return
eclampsia the maternal response to the oxidatively damaged to homeostasis. ER stress in the liver is particularly sensitive to
placenta is what could have been predicted and what has inflammatory stimuli and underlies the acute-phase response.
been found. The systemic inflammatory response has mas-
sive and wide-ranging consequences, all of which are seen WIDESPREAD IMPLICATIONS OF VASCULAR
in the second stage of preeclampsia.19 The nature of the vas-
INFLAMMATION
cular inflammation has unique attributes induced by release
of factors from the placenta which are not present in non-
It is self-evident that vascular inflammation involves endo-
pregnant individuals. These include the angiogenic placental
thelium as well as inflammatory leukocytes (granulocytes,
growth factor (PlGF) and the antiangiogenic factors, soluble
macrophages, and natural killer lymphocytes). However,
vascular endothelial growth factor receptor-1 (sVEGFR-1)
the coagulation system, liver and adipose tissue also directly
and soluble endoglin (sEng) as described in Chapter 6.
contribute soluble factors to the inflammatory response
(Table 8.3). The full extent of vascular inflammation on
diverse systems may not be appreciated, nor the two-way
ENDOTHELIAL CELLS ARE
interactions between its components. For example, blood
INFLAMMATORY CELLS
coagulation is not only activated by inflammatory processes
but thrombin, the final trigger to coagulation, also stimulates
It has long been known that preeclampsia can be largely
inflammation via specific receptors. Angiogenesis,37 oxida-
explained by diffuse maternal endothelial cell dysfunc-
tive stress,38 and obesity39 are all tightly linked to inflamma-
tion34 (see Chapter  9). In the circulation, endothelial cells
tory responses and all highly relevant to preeclampsia, as is
are key players in systemic inflammatory responses as
described subsequently. The acute-phase response is a com-
well as mediating local inflammation by upregulation of
plex inflammatory stress response originating from the liver.
adhesion molecules that tether and then anchor margin-
Nuclear factor-κB (NF-κB) is a transcription fac-
ated leukocytes. In the field of atherosclerosis this is well
tor expressed by nearly all cells, which is central to many
researched. Atherosclerosis is a focal large-vessel dis-
inflammatory responses. It is activated by numerous stress-
ease; whereas microvascular endothelium promotes diffuse
ors, including inflammatory and oxidative stresses, and
systemic inflammation.35 Both are examples of vascular
suppressed by hCG, estrogen, IL-4, and IL-10.40 It is also
inflammation. If preeclampsia is an endothelial disorder
activated by oxidative and endoplasmic reticulum (ER)
then the corollary is that it is also an inflammatory disorder.
stress.41,42 These stresses also are prominent features of
The latter is a more generalized concept that subsumes the
the syncytium in the preeclampsia placenta.33 The interac-
former. Inflammation is one part of a more generalized and
tion between NF-κB and hypoxia inducible factor (HIF)-1α,
integrated stress response, hence other systems are inevita-
the transcription factor that controls cellular responses to
bly involved and the stresses distributed between different
low oxygen tension, is particularly close. HIF-1α activates
cellular systems. Cellular oxidative stress will for example
transcription of over 400 genes involved in the regulation of
induce endoplasmic reticulum (ER) stress, the unfolded pro-
immune and inflammatory responses and related functions.
tein response and an inflammatory response (reviewed in19).
The acute-phase response is a complex inflammatory stress
response originating from the liver.
INFLAMMATION AND THE INTEGRATED
STRESS RESPONSE TABLE 8.3  Components of the Inflammatory or
Innate Immune System
The integrated stress response (ISR) is evolutionarily Inflammatory leukocytes:
ancient.36 Central to the response is ER stress and the  Granulocytes
unfolded protein response, which enhances removal of  Monocytes
unfolded proteins and re-programs protein translation in   Natural killer lymphocytes
the ER. While most protein synthesis is reduced, that of   Certain B cells producing “natural antibodies”
specific transcription factors, promoting production of Endothelium
stress response proteins that restore homeostasis, is aug- Platelets
mented via numerous sensors of cell damage or dysfunc- Coagulation cascade
tion. Among the stress response proteins are molecular Complement system
Cytokines and chemokines
chaperones, antioxidants, transcription factors and so on.
Adipocytes
Inflammation is part of the ISR; it activates and is activated Hepatocytes
by oxidative stress and ER stress.36
Chapter 8 · Immunology of Normal Pregnancy and Preeclampsia 167

Vascular inflammation brings in its trail the secretion of been known for many years (see review by Harris et al.).50
many factors that orchestrate its expression. Many of these Lipolysis releases free fatty acids, which contribute to the
are altered in preeclampsia. insulin resistance peripherally.
The origins of preeclampsia lie in the placenta, specifi- Obesity, which is a risk factor for preeclampsia
cally the syncytium, which demonstrates all the indicators (Chapter  7), is characterized by a vascular inflammatory
of an ISR.33 Three major stresses – ER, inflammatory and response. It arises because adipose tissue is not simply an
oxidative – are particularly relevant to preeclampsia and energy store but a source of proinflammatory cytokines and
tightly interlinked. A number of trophoblast-derived media- other metabolic mediators (adipokines) as mentioned in
tors, discussed below and in Chapter  6 (sVEGR-1, PlGF), the preceding section and is the principal source of leptin.
disseminate the placental problem by causing vascular Leptin is secreted during acute inflammation and has an
inflammation, which provokes widespread dysfunction in important action on immune cells, all of which express
many maternal systems. Ultimately, the dysfunction is more the leptin receptor. Leptin thereby causes or enhances pro-
than simply vascular and has major metabolic effects.43 inflammatory responses, as reviewed by Matarese et  al.51
It can be classified as a cytokine as well as an adipokine.
The net effect is that obesity is a state of chronic systemic
CYTOKINES, CHEMOKINES, GROWTH inflammation.
FACTORS, ADIPOKINES AND The importance of obesity in generating the inflamma-
ANGIOGENIC FACTORS tory response is demonstrated by its reversal after weight
loss. Visceral rather than subcutaneous fat is more important
A diverse group of secreted proteins and glycoproteins coor- in this context. Obesity is a key part of the metabolic syn-
dinate the inflammatory response. The term cytokine origi- drome,52 which also includes insulin resistance with impaired
nally referred to peptides that are produced by and act on glucose tolerance or overt diabetes, dyslipidemia, and hyper-
immune and hematopoietic cells. Although they are criti- tension. Obesity-associated IL-6 can induce the acute-phase
cal for both innate and adaptive immune function, they can response including production of circulating C-reactive pro-
also be secreted by non-immune cells and have non-immune tein (CRP). Elevated CRP is a typical feature of the meta-
functions that are relevant to the pathophysiology of pre- bolic syndrome but other plasma acute-phase reactants, such
eclampsia. Some are chemokines, for example interleukin-8 as fibrinogen, are similarly increased. Some acute-phase
(IL-8), that stimulate the migration of inflammatory cells. proteins are listed in Table 8.4. In the mouse liver the acute-
Adipokines include proteins that are secreted from (and phase response involves nearly 10% of the genome,53 indicat-
synthesized by) adipocytes but exclude products of other ing the complexity and magnitude of the response.
cell types in adipose tissue, such as macrophages.44 They Vascular inflammation is accompanied by an increase
include cytokines such as IL-6, classical adipokines (leptin, in triglyceride-rich lipoproteins, a reduction in high-den-
resistin, adiponectin) and even acute-phase proteins (PAI-1, sity lipoprotein cholesterol, and impairment of cholesterol
angiotensinogen). The classical adipokines all have actions transport. These metabolic alterations, which promote ath-
on immune cells, that is they have cytokine-like activity, as erosclerosis, may explain an epidemiological link between
well as angiogenic activity, as reviewed by Ribatti et  al.45 chronic inflammation and cardiovascular disease.54
Angiogenic factors (see Chapter  6) include VEGF, which The term metaflammation has been applied to the com-
is also a growth factor for endothelium. Other biologically bination of low-grade chronic vascular inflammation and
active proteins or peptides can have cytokine-like activity, metabolic changes. It underlies chronic conditions such as
such as angiotensin II (Ang II); which is directly proinflam-
matory.46 By its induction of VEGF, Ang II is also indirectly
angiogenic.47 Insulin on the other hand, while it is a mito- TABLE 8.4  Changes in Concentrations of Plasma
gen, is antiinflammatory, as reviewed by Dandona et  al.48 Proteins in the Acute-Phase Response
Despite these cytokine-like actions, Ang II and insulin are Positive acute-phase reactants, increased in the circulation
not considered to be cytokines. In summary, the nomencla- ●
CRP (C-reactive protein)
ture surrounding these factors can be confusing and limit an ●
Angiotensinogen
understanding of their wider actions and their involvement ●
Fibrinogen, prothrombin, Factor VIII, plasminogen
with inflammatory responses in particular. ●
Complement components: (eg C3)

Alpha-1-antitrypsin, alpha-1-antichymotrypsin

Haptoglobin, hemopexin, ceruloplasmin
METABOLISM AND VASCULAR ●
Soluble phospholipase A2
INFLAMMATION ●
Sialic acid, α1-acid glycoprotein
Negative acute-phase reactants, reduced in the circulation
Circulating proinflammatory factors such as endotoxin or

Albumin

Transferrin
tumor necrosis factor (TNF)-α cause insulin resistance and ●
Retinol-binding protein
hence hyperlipidemia.49 The hyperlipemia of sepsis has
168 Chesley’s Hypertensive Disorders in Pregnancy

obesity, atherosclerosis and type 2 diabetes,55 and, as dis- TABLE 8.5  The Vascular Inflammatory Network
cussed above, shares many features with preeclampsia. is Stimulated in Normal Pregnancy Relative
to Non-Pregnancy
*Leukocytosis66
ACUTE-PHASE RESPONSE *Increased leukocyte activation64
*Complement activation67
The acute-phase response may also be a chronic response to *Activation of the clotting system68 See Chapter 17
*Activation of platelets69 See Chapter 17
local or systemic inflammation. It comprises variable changes
*Markers of endothelial activation70 See Chapters 9 and 17
in circulating plasma protein concentrations and other phe-
*
nomena such as fever, anemia, leukocytosis and metabolic Significant change(s) relative to normal non-pregnant women.
adaptations especially involving the liver and adipose tis- Not all authors agree; see text. There are usually multiple references to
justify each change.
sue.56 Proteins linked to this response, acute-phase proteins,
are synthesized in the liver. They are classified as positive if
they increase with systemic inflammation (e.g., C-reactive
protein, CRP) or negative if they decrease (e.g., albumin)
TABLE 8.6  The Systemic Inflammatory Network is
(Table 8.4). Stimulated in Preeclampsia Relative to Normal Pregnancy
The concentrations of CRP increase rapidly in response
to inflammatory stimuli. Human CRP binds with high *Leukocytosis77
affinity to phosphocholine residues and other intrinsic and *Increased leukocyte activation64
§
extrinsic ligands, including native and modified plasma lipo- *Complement activation78
*Activation of the clotting system See Chapter 17
proteins, damaged cell membranes and various phospholip-
*Activation of platelets See Chapter 17
ids and constituents of microorganisms and plant products.
*Markers of endothelial activation See Chapters 9 and 17
CRP is therefore a pattern-recognition receptor for a range Increased circulating proinflammatory
of “danger” molecules.57 cytokines
  * Plasma tumor necrosis factor-α65
  §*Plasma interleukin-679
  §*Plasma interleukin-880
VASCULAR INFLAMMATION IN NORMAL
PREGNANCY AND PREECLAMPSIA *
§
Significant change(s) relative to normal pregnant women.
Not all authors agree; see text.
There are usually multiple references to justify each change
A subtle systemic inflammatory response precedes con-
ception in the luteal phase of the menstrual cycle58 but
becomes overt in the first trimester of pregnancy. Features
of the response are wide-ranging. Some that are deemed to a converse fashion, oxidative stress can stimulate an inflam-
be physiological adaptations of pregnancy are in fact com- matory response.74
ponents of the acute-phase response such as reduced plasma Measurements of circulating inflammatory cytokines
albumin59 and increased fibrinogen concentrations.60 Many during pregnancy can give complex results, owing in part to
such examples have been summarized by Redman and circulating binding proteins. Direct measurement of plasma
Sargent.61 CRP is modestly elevated in pregnancy, starting concentrations of the proinflammatory cytokines interleukin-6
in the first trimester,62 when the long-recognized leukocyto- (IL-6) and tumor necrosis factor-α (TNF-α) show increased
sis of pregnancy, another sign of systemic maternal inflam- levels relative to non-pregnancy towards the end of gesta-
mation, is established.63 tion. Another method is to measure their production within
As pregnancy advances the vascular inflammation strength- peripheral blood mononuclear cells ex vivo, before or after
ens to peak during the third trimester. The concept was consoli- stimulation. Intracellular cytokines can be measured flow
dated by flow cytometric analyses of circulating inflammatory cytometrically, but the intense stimulation required can intro-
leukocytes.32,50,64 The changes are associated with increases in duce artifacts. Studies of peripheral blood cells exclude the
circulating inflammatory cytokines in the second half of preg- possible contributions of endothelial cells which secrete IL-6,
nancy (several reports, cited in65; Table 8.5). TNF-α and several other cytokines, as well as chemokines.75
The inflammatory changes are associated with evidence Stage 2 preeclampsia, which appears to originate from
of increasing systemic oxidative stress, in terms of several the syncytial surface (Interface 2) of the placenta, is char-
circulating markers particularly oxidized lipids.71,72 In this acterized by an exaggerated maternal vascular inflammatory
regard, it is key that oxidative stress and chronic inflam- response involving the inflammatory biomarkers of nor-
mation are related processes in ISR.15 The inflammatory mal pregnancy,76 which are more severely affected in pre-
response generates oxidative stress (reviewed in73) and, in eclampsia (Table 8.6).
Chapter 8 · Immunology of Normal Pregnancy and Preeclampsia 169

THE CONTINUUM BETWEEN NORMAL IFN-γ and IL-2 are produced by Th1 cells. Cytokines
PREGNANCY AND PREECLAMPSIA that are derived from monocyte/macrophages or other
sources are more likely to be stimulated by the systemic
The systemic inflammatory response of preeclampsia is inflammation of normal pregnancy. For example, preg-
not a unique condition but represents the extreme part of a nancy peripheral blood monocytes are primed to produce
spectrum that is common to all pregnancies. Preeclampsia more of the type 1 cytokine, IL-12, than non-pregnancy
develops when vascular inflammation overcomes protec- monocytes.87 IL-12 is a potent stimulator of other Type 1
tive maternal compensation.32 Because of the continuum, responses (IFN-γ production), which implies that strong
preeclampsia is clinically distinguished by arbitrary and specific stimuli might rapidly convert the Type 2 bias of
artifactual thresholds. This explains why preeclampsia has normal pregnancy to Type 1. Hence, the inflammatory
evaded diagnostic categories or identification of clear path- system in pregnancy can be said to be meta-stable. It is
ological lesions distinct from normal pregnancy. This in restrained from producing Type 1 cytokines, but is primed
turn has important implications for prediction, screening, to do so if it is challenged.
and studies of genetic susceptibility or treatment.32 The Th1/Th2 paradigm can be extended to include
Th17.81 In general Th subsets inhibit each other: IFN-γ
inhibits proliferation of Th2 cells; conversely IL-4 from
IMMUNOREGULATION Th2 cells inhibits expansion of Th1 cells. IL-17 from Th17
cells suppresses Th1 differentiation, whereas both Th1 and
Inflammation stimulates adaptive immunity, which requires Th2 inhibit Th17.88
modulation to minimize potentially damaging transplan- Th17 and Tregs are very closely related and therefore
tation reactions directed against the antigenically foreign are considered together.
fetus. During pregnancy, immunoregulation affects the
gamut of T helper cell activity, namely Th1, Th2, Th17,
memory T, and T regulatory cells (Tregs). There are other T REGULATORY CELLS, TH17
changes associated with preeclampsia described below.
AND T-CELL MEMORY
T helper (Th) cells are ubiquitous lymphocytes that
have no direct actions against pathogens, tumor cells or
transplanted cells. Instead they direct other lymphocytes to
Normal Pregnancy
deliver antigen-specific effector responses; via either cyto- Tregs are a heterogeneous class of suppressor T cells,
toxic (T cells) or antibody-producing B cells. T helper cells which share the transcription factor Foxp3 but lack other
secrete cytokines and related proteins that interact with specific markers. They are antigen specific and suppress
other leukocytes. Subsets of Th cells orchestrate different activation of cytotoxic T cells but may stimulate B cells, for
patterns of signals to achieve distinct activities (reviewed example to produce IgA. They may be thymically derived
by Basu et al.81). Th1 cells promote cell-mediated immunity natural Tregs (nTregs) or induced iTregs. iTregs suppress T
and secrete “Type 1” cytokines such as interleukin-2 (IL-2), cell differentiation in both antigen-specific and nonspecific
and interferon-γ (IFN-γ); Th2 cells promote humoral immu- ways.88
nity via “Type 2” cytokines such as IL-4, IL-5, IL-10, and Tregs suppress other T cells, NK cells, macrophages
IL-13. A third subset of IL-17-producing Th17 cells has and dendritic cells.89 They restrain autoimmunity and are
been added to this list. Th17 cells share programs of differ- the primary mediators of tolerance, for which reason they
entiation with naïve T cells and T regulatory cells (Tregs) are highly relevant to the immunology of pregnancy, for
and are discussed below. Other subsets, Th3 and Th22, are example, essential for the success of murine pregnancy.90
less well defined and not considered here. Tregs act in tissues by direct cell contact, hence, their con-
For many years normal pregnancy has been associated centrations in peripheral blood at best are only indirectly
with the concept of a Th2 bias in immune function82 char- informative. Of interest is that their levels are stimulated by
acterized by reduced lymphocyte secretion of IFN-γ83 and estrogens, with increases in both the follicular phase of the
IL-2,84 and increased production of IL-4 and IL-10. menstrual cycle91 and early pregnancy.92
The same shift is evident in decidual lymphocytes85 Tregs in the decidua are probably the most relevant,
and in circulating NK and NKT cells, but not detectable in where they are enriched relative to concentrations in
unstimulated T cells,86 leading to the suggestion that NK peripheral blood.93 It is speculated that decidual Tregs
or NKT cells may be the first to impose this bias in preg- downregulate maternal responses to HLA-C-incompatible
nancy.86 The Th2 cytokine balance is superimposed on the extravillous cytotrophoblast.94 This is a very important con-
normal innate immune stimulation of pregnancy, restrain- cept; if confirmed, it points to the involvement of T cells in
ing and redirecting lymphocyte activity toward a systemic partner-specific immunity, the involvement of T cell mem-
humoral inflammatory response. ory, and a potential mechanism for the second pregnancy
170 Chesley’s Hypertensive Disorders in Pregnancy

phenomenon of preeclampsia. To date, in preeclampsia, ANGIOTENSIN II (ANG II), THE IMMUNE


only peripheral blood Tregs have been studied. The results SYSTEM AND PREECLAMPSIA
are inconsistent: both reduced95 and unchanged96 numbers
have been reported. However, the importance of Tregs, par- Ang II is a classical mediator of hypertension and of salt
ticularly those residing at Interface 1 in Stage 1 preeclamp- and fluid retention. It is also an important contributor to
sia, has not been adequately assessed. Here, they would be hypertensive end-organ damage. Its pathophysiology in
predicted to play an important role in the control of placen- preeclampsia is discussed extensively in Chapter  15. As
tation. Tregs have a reciprocal relationship with Th17 cells, already mentioned, in addition to its vasoactive proper-
the latter promoting autoimmunity.97 Both T cell subtypes ties, Ang II is potently proinflammatory. It activates NF-κB
require TGFβ for differentiation from naïve T cells, and through the AT1R (angiotensin II type 1 receptor) to
therefore share many early features in their transcriptional induce inflammatory cytokines, chemokines, and growth
programs. Pathogenic Th17 cells, in addition, need IL-6. factors that mediate the tissue damage.106 In fact, Ang II
High concentrations of TGFβ favor Treg differentiation affects all aspects of immunity, innate and adaptive, cellu-
whereas lower concentrations combined with IL-6 promote lar and humoral.107 For example T cells and NK cells both
Th17 cell production.81 Both Th1 and Th2 cells inhibit their have their own functional renin-angiotensin systems.108
development. There is no change in the proportion of circu- Furthermore at least some of the hypertension induced by
lating Th17 lymphocytes during normal pregnancy.98 Ang II depends on its direct action on T cells. An indirect
A key feature of Tregs and Th17 cells is their plastic- effect, possibly involving Ang II stimulation of dendritic
ity.88 Th17 cells can transform into Th1 cells, while Tregs cells, may also contribute. The specific T cells involved are
can transform into Th17, although the converse does not atypical, being negative for CD4 and CD8, and represent an
happen.88 Thus the Th biases or states that are typical of inflammatory subtype that produces IL-17.109
normal pregnancy or preeclampsia are dynamic. In preeclampsia the renin-angiotensin system is typi-
Preeclampsia has been associated with a Th183 and cally suppressed relative to normal pregnancy.110 But an
Th1799 bias in peripheral blood lymphocytes with a relative alternative ligand of ATR1 is its agonistic autoantibody
deficiency in Tregs.100,101 Since Th17 cells are promoted by (AT-AA), which is highly associated with the disorder111
lower availability of TGFβ it is possible that circulating sEng and potentially contributes to its hypertension and other
(a TGFβ-binding protein; see Chapter  6), interferes with features (see Chapter  15). It represents a more general
T cell differentiation. TGFβ is proposed to promote differen- phenomenon of autoantibodies (agonistic or antagonis-
tiation of Th17 cells at the expense of Tregs, contributing to tic) against G-protein-coupled receptors, for example TSH
a proinflammatory environment with increased IL-17.99 receptor antibodies in relation to Graves disease.112
In preeclampsia the specific Type 2 bias of normal As indicated above, a Th17 bias, such as occurs in pre-
pregnancy is lost with increased production of IFN-γ. and eclampsia, would be expected to stimulate autoimmunity. In
IL-2.71,72 Circulating IL-17 concentrations appear to be addition experimental, Ang II-induced hypertension is not
unchanged102 despite an increase in Th17 cell counts. There sustained in IL-17(−/−) mice,113 indicating possible par-
are increases in a range of other cytokines, chemokines, ticipation of IL-17 not only before but after the appearance
adipokines and antiangiogenic factors, which all reflect an of such antibodies. But the precise mechanisms of the gen-
inflammatory response. Circulating adipokines with cyto- eration of this antibody are not known. Its activity declines
kine activity, either proinflammatory (leptin51) or antiin- relatively quickly (within the first two weeks after preg-
flammatory (adiponectin103) activities are also increased in nancy) but does not disappear111 and persists to cause recur-
preeclampsia. rent problems in further pregnancies. Hence AT-AA would
Finally, the central issue for this chapter is whether be predicted to cause recurrent problems in later pregnan-
partner-specific memory develops and persists to enhance cies, as do antiphospholipid autoantibodies. It is not known
maternal immune tolerance in the next pregnancy by the whether AT-AA are present before conception of eventu-
same partner. Whereas such memory could be associated ally preeclamptic pregnancies, but their persistence would
with NK cells, whose responses to specific stimuli can be not be consistent with the first-pregnancy preponderance of
“educated” or “licensed,”104 memory is classically associ- preeclampsia.
ated with T cells. In relation to pregnancy, what needs to
be sought is partner-specific memory carried by Tregs.
There is no evidence for such in humans but there is in SYSTEMIC IMMUNOREGULATION IN
mice. Mating-specific Tregs accumulate in the secondary NORMAL PREGNANCY AND PREECLAMPSIA
lymphoid tissues of pregnant mice, persist after pregnancy,
and give an accelerated response in a second pregnancy. If a Humoral factors also contribute to immunoregulation dur-
similar mechanism applied to human pregnancy this would ing normal pregnancy. Indoleamine 2,3-dioxygenase (IDO)
be the most likely explanation for the “preeclampsia second is a widely distributed enzyme that catabolizes tryptophan.
pregnancy effect.”105 It is characterized best in monocytes and macrophages but
Chapter 8 · Immunology of Normal Pregnancy and Preeclampsia 171

can be produced by other cell types, such as endothelial arteries, characterized by lipid deposition and oxidative
cells. It is primarily induced by IFN-γ, but other proinflam- stress.123 The trigger seems to be chronic, focal endothelial
matory stimulants are also effective. By reducing the avail- activation from the shear stress of turbulent blood flow124 or
ability of tryptophan, indoleamine 2,3-dioxygenase inhibits other undefined factors. Enhanced focal endothelial adhe-
immune cell function (summarized in95). IDO is reduced in siveness attracts immune cells, including monocyte/macro-
preeclampsia as judged by direct and indirect measures in phages, to the subendothelium, where increased endothelial
the placenta or indirect measures of maternal plasma.95 The permeability enables lipoproteins, with their cargo of lipids,
effect would be to diminish the inhibitory action of trypto- to be scavenged by macrophages that transform into foam
phan depletion, resulting in immune cell activation. cells. Foam cells are not specific to atherosclerosis, but
HLA-G is a non-classical HLA molecule expressed form in other inflammatory states, such as tuberculomas or
almost exclusively by non-villous trophoblast. Unlike graft rejection (see below).
classical HLA molecules it has limited polymorphism Other than macrophages, dendritic, T and B cells, as well
(reviewed by Apps et al.18). It is immunosuppressive rather as NKT cells and possibly NK cells contribute to matura-
than stimulatory (reviewed by Sargent96) by way of its tion of atherosclerosis. Th1 cells are proatherogenic, while
interactions with receptors expressed by NK cells, antigen- subsets of T regulatory cells (Tregs) are protective (reviewed
presenting cells and some T cells.102 Expression of HLA-G by122). This implies that specific antigens are important.
by extravillous cytotrophoblast is reduced in preeclamp- Atherosclerosis has a longer time course, with terminal
sia114 and may contribute by undefined immune mecha- plaque formation and rupture. Acute atherosis affects only
nisms to the associated restricted invasion of spiral arteries small spiral arteries as does the atherosis of graft vascular
in the placental bed (see Chapter  5). Soluble HLA-G is disease (GVD), which complicates the integrity of solid
released by protelolytic cleavage of the membrane-bound allografts (such as of the kidney or heart), even when donors
protein.115 It is antiangiogenic116 and appears to be immu- and recipients are HLA-compatible.125 GVD depends on
nosuppressive116 and is reduced in preeclampsia,117 con- both innate and adaptive immune responses.125 The similari-
sistent with the view that downregulation of the immune ties between acute atherosis and GVD have been highlighted
system is impaired in this condition. previously and include fibrinoid necrosis (medial smooth
muscle cells), intimal hyperplasia, and foam cell forma-
tion.126 Intimal hyperplasia is more prominent in GVD125
ACUTE ATHEROSIS: A SECOND than in acute atherosis. Both lesions involve perivascular
INFLAMMATORY LESION lymphocyte infiltration, complement and immunoglobulin
OF PREECLAMPSIA deposits and occur at the boundaries between tissues from
genetically different individuals. Both forms of atherosis are
Acute atherosis is a uteroplacental spiral artery lesion, char- associated with conditions where there are circulating ago-
acterized by subendothelial lipid-filled foam cells, fibrinoid nistic AT-AA.127
necrosis and leukocyte infiltration.118 The foam cells are Whatever its cause it is reasonable to conclude that
predominantly CD68-positive macrophages. Acute athero- acute atherosis is a secondary consequence of the vascular
sis most often occurs distally, downstream of inadequately inflammation that occurs in preeclampsia and even nor-
remodeled spiral arteries in the myometrium, namely at the mal pregnancy and may reflect a true, low-grade maternal
fully remodeled tips of their decidual segments. The lesion immune rejection of the fetus. Its development is likely
is focal, not necessarily affecting all spiral arteries or their to involve maternal specific constitutional factors. By its
entire length. The time course of acute atherosis is not well impact on uteroplacental perfusion it adds a further stage to
defined. It appears to regress after delivery. the pathogenesis of preeclampsia, affecting a subset of all
Its incidence depends on patient selection and tissue mothers (Fig. 8.4).
sampling.119 It affects 20–40% of preeclamptic women and
is associated with more severe forms of the disorder.120 It
also occurs without preeclampsia – in normotensive fetal THE ROLE OF THE PLACENTA
growth restriction or with underlying medical problems AND NON-PLACENTAL FACTORS
such as diabetes mellitus. It is an incidental finding in up to
15% of normal pregnancies.121 Since the placenta is ultimately the cause of preeclampsia
The arterial wall lesion can reduce spiral arterial cali- (see Chapter 5) it would be predicted that one or more fac-
ber and blood flow, which would be expected to exacerbate tors released from the syncytial surface of the placenta into
placental dysfunction and oxidative stress especially as it the maternal circulation are the proinflammatory causes.
is associated with spiral arterial thrombosis and placental It is possible that syncytiotrophoblast could be a direct
infarction (reviewed in121,122). source of proinflammatory cytokines. Of most interest
Acute atherosis resembles atherosclerosis, which is a is secretion of VEGF, which is not only angiogenic, but
chronic inflammatory lesion of large and medium-sized strongly proinflammatory128; this is to be expected since
172 Chesley’s Hypertensive Disorders in Pregnancy

Four stages of preeclampsia

Stage 1 Poor maternal tolerization to


paternal antigen by semen exposure No symptoms
Pre-conception

Stage 2 Poor placentation No symptoms


First half of pregnancy

Stage 3 Oxidatively stressed Incipient preeclampsia


Second half of pregnancy placenta

sFlt-1 and other mediators

Dysfunctional maternal endothelium Overt preeclampsia


clinical signs of preeclampsia

Acute atherosis
Stage 4
Deterioration of placental perfusion Severe preeclampsia
End pregnancy
Placental infarction

FIGURE 8.4  Two further stages are added to this adaptation of the two-stage model. Pre-conceptual maternal tolerization to partner’s
antigens is achieved by pre-conceptual exposure to paternal semen (new stage 1). Stages 2 and 3 are, respectively, stages 1 and 2 of the
original two-stage model. Stage 4 comprises the development of acute atherosis in the spiral arteries which causes further adverse effects
on uteroplacental perfusion, particularly from thrombosis. Acute atherosis probably affects less than half of preeclampsia cases. (This figure
is reproduced in color in the color plate section.)

angiogenesis is driven by inflammatory mechanisms. VEGF cause oxidative stress, reducing the bioavailability of nitric
is strongly expressed in syncytiotrophoblast and upregu- oxide by uncoupling its synthesis. In this context dys-
lated by hypoxia. Its circulating levels are measured as regulation of nitric oxide availability ascribable to VEGF
either higher or lower than normal in preeclampsia: total antagonism is likely to be only one part of a bigger picture
VEGF is increased, but free VEGF is reduced, because in which oxidative and inflammatory stresses are critical.
it induces, and is bound to, a circulating soluble receptor The proximal cause of sVEGFR-1-induced hypertension
(sVEGFR-1 or sFlt-1). sVEGFR-1 is also produced by the is endothelin,132 a classical vasoconstrictor and inflam-
placenta (see Chapter 6). matory mediator. Circulating endothelin-1 is increased
Whereas sVEGFR-1 is antiinflammatory in other medi- in preeclampsia relative to normal pregnancy.132,133 It is
cal contexts,129 sVEGFR-1 release from trophoblast130 and worth remembering that most endothelin is produced from
macrophages129 is enhanced by inflammatory stimuli. While the abluminal aspect of endothelial cells134 where it reacts
hypoxia has previously been given prominence as the trigger locally with vascular smooth muscle. So circulating levels
for release from the preeclampsia placenta (Chapter 6), inflam- are unlikely to be more than distant reflections of important
matory mechanisms may contribute or even predominate, constrictor mechanisms. Paradoxically, altlhough sVEGF-
especially as the activation of the hypoxia response requires R1 might be expected to be antiinflammatory because it is
the action of the NF-κB. The latter transcriptionally regulates antiangiogenic, in the context of oxidative stress it becomes
stress responses, including those mediated by HIF-1α.74 proinflammatory because it amplifies vascular NO defi-
VEGF-R1 is also the primary receptor for placental ciency in endothelium and/or vascular smooth muscle.
growth factor (PlGF), which is another cytokine and angio- There are several other circulating factors derived from
genic factor produced by the placenta. Its circulating con- syncytiotrophoblast that could affect systemic inflammation
centrations are reduced in preeclampsia (see Chapter 6). One or angiogenesis or both. They are summarized in Table 8.7.
relevant function of PlGF is, like VEGF, proinflammatory. It Note that their circulating levels are increased in normal
can activate monocytes and increase mRNA levels of proin- pregnancy and in, most instances, increased further in pre-
flammatory cytokines (TNF-α, IL-1β) and chemokines.131 eclampsia, consistent with the hypothesis that pregnancy
However, in that PlGF production is diminished, it does not is an intermediate state between non-pregnancy and pre-
appear to be a placental factor contributing to the excessive eclampsia. Note also that they include not only classical
inflammatory response of preeclampsia. cytokines, but growth factors and adipokines. Their rela-
Agonistic AT-R1 autoantibodies would be expected, tive contributions to systemic inflammation in normal preg-
as does Ang II. to stimulate vascular NADPH oxidase and nancy have yet to be defined.
Chapter 8 · Immunology of Normal Pregnancy and Preeclampsia 173

TABLE 8.7  Inflammatory Responses and Factors Secreted by Syncytiotrophoblast

Factor Secreted by *Pro (↑) or Anti (↓) Normal Pregnancy Relative to Preeclampsia
Trophoblast Inflammatory Non-Pregnancy Relative to Normal
Pregnancy

PlGF ✓Chapter 6 ↑130 ↑Chapter 6 ↓Chapter 6


sVEGFR-1 ✓Chapter 6 Variable ↑Chapter 6 ↑Chapter 6
s-Endoglin ✓Chapter 6 Not defined ↑Chapter 6 ↑Chapter 6
Activin-A ✓135 ↑136 ↑137 ↑138
Inhibin-A ✓135 ↓136 ↑137 ↑138
CRH ✓139 ↑ circulating38 ↑140 ↑140
Leptin ✓141 ↑51 ↑141 ↑142

TROPHOBLAST EXTRACELLULAR TABLE 8.8  Circulating Microvesicles in Healthy


VESICLES Non-Pregnant Individuals (n =15)
Platelets 237 × 106/L
Cell membrane microvesicles are shed during apopto- Endothelial cells 64 × 106/L
sis or cell activation. In healthy individuals, microvesicles Granulocytes 46 × 106/L
from platelets, endothelium, neutrophils, and erythrocytes Erythrocytes 28 × 106/L
normally circulate, with platelet microvesicles being the
Berckmans et al. 2001.143
most abundant. In inflammatory conditions they increase. Constitute 5–50 μg protein/mL of plasma.
Their concentrations are listed in Table 8.8. Microvesicles
may be proinflammatory, antiinflammatory or procoagu-
lant (reviewed by Redman and Sargent144). Procoagulant for their cargo of microRNAs, which are small non-cod-
platelet-derived microvesicles (platelet dust) can dissemi- ing molecules that regulate gene expression, transcrip-
nate and amplify localized clotting. The syncytial surface tionally and post-transcriptionally. Little is known about
of the placenta is a transitory intrusion into the maternal differences in circulating exosomes in preeclamptic rela-
circulation, where it sheds a wide range of cellular and tive to normal pregnancies. miRNAs are emerging that are
subcellular debris, including syncytiotrophoblast microves- biomarkers for preeclampsia, for example, miR-210,151
icles (reviewed in145). We have proposed that clearance of but none that have been linked so far to inflammatory or
the debris provokes a systemic inflammatory stimulus in immune functions.
normal and preeclamptic pregnancies.
Syncytiotrophoblast microvesicles are shed in signifi-
cantly increased amounts in preeclampsia, but not in nor- MATERNAL PREDISPOSING FACTORS
motensive intrauterine growth restriction.146 They have a
profound antiendothelial effect147 and are also proinflam- Some medical conditions are well known to predispose
matory in vitro.148 to preeclampsia, including obesity, diabetes, and chronic
Shedding of debris from the syncytial surface would hypertension. The confluence of these three signs is
be expected to increase in two situations. The first is with referrred to as the metabolic syndrome and its relationship
increased placental size. Preeclampsia is predominantly a with preeclampsia is explored extensively in Chapter 7. The
disorder of the third trimester, when the placenta reaches its effect of these medical conditions is to elevate the prepreg-
greatest size. The prevalence of preeclampsia also increases nancy baseline of systemic inflammation upon which the
with multi-fetal pregnancies and with placental oxida- changes of pregnancy are superimposed (Fig. 8.5). We pro-
tive stress. Severe preeclampsia, typically of early onset, pose that, in pregnancy complicated by such conditions, the
is associated with abnormally small placentas and intense decompensation from excessive systemic inflammation will
fetal growth restriction. These are likely to be affected happen earlier, accounting for the predisposition of affected
by an alteration in the quality of inflammatory stimuli, women to preeclampsia. It will also persist from pregnancy
for example, increased content of peroxidized placental to pregnancy, leading to recurrent preeclampsia.
lipids.149 Low-grade systemic inflammation is also a feature when
The maternal circulation also contains trophoblast- obesity,152 diabetes,153 and chronic hypertension154 coex-
derived nanovesicles, more correctly termed exosomes. ist in men or non-pregnant women. Thus, it is not surprising
These are too small (~100 nm) to be reliably detected that the metabolic syndrome predisposes to preeclampsia.154
by conventional flow cytometry.150 They are considered Indeed the metabolic effects of pregnancy, with or without
to be physiological intercellular signals and are notable preeclampsia, have been likened to this syndrome.155,156
174 Chesley’s Hypertensive Disorders in Pregnancy

‘Placental’ preeclampsia ‘Maternal’ preeclampsia FIGURE 8.5  Systemic inflammation in preg-


nancy with and without conditions that cause a
chronic systemic inflammatory response such as
Normal
Preeclampsia Severe inflammatory diabetes, obesity, chronic hypertension or chronic
placenta
Abnormal placenta dysfunction with maternal infection. These conditions predispose
but signs of
clinical signs preeclampsia to preeclampsia as the inflammation of pregnancy
starts from a higher baseline. Because of this they
also predispose to preeclampsia in subsequent
Increasing systemic pregnancies.
inflammatory Non-
Normal stress pregnancy
pregnancy abnormal women
with higher baseline

Non-
pregnancy

Preeclampsia, vascular inflammation and its consequences


Anti-AT1 receptor
antibodies

Inflammatory Activated
leukocytes Endothelium Increased autoimmunity

Th1, Th17

Vascular inflammatory stress

Liver Th2, Tregs

Adipose tissue Clotting Platelets Complement

Acute phase response

Insulin resistance Dyslipidemia


FIGURE 8.6  Vascular inflammation involves not only inflammatory leukocytes, but the endothelium, platelets, clotting and comple-
ment systems. Metabolic changes lead to insulin resistance and dyslipidemia. The inflammatory stress deviates away from the Th2 bias of
normal pregnancy towards Th1/Th17. Th17 and loss of Tregs predispose to autoimmunity and may help stimulate formation of anti-AT1
receptor autoantibodies. These are agonistic and therefore proinflammatory. These processes contribute towards many recognized features
of preeclampsia. (This figure is reproduced in color in the color plate section.)
Chapter 8 · Immunology of Normal Pregnancy and Preeclampsia 175

CONCLUSIONS 3. Robillard PY, Hulsey TC, Alexander GR, Keenan A, de


Caunes F, Papiernik E. Paternity patterns and risk of pre-
Normal pregnancy involves two broad aspects of mater- eclampsia in the last pregnancy in multiparae. J Reprod
nal immune function: adaptive immunity, with recognition Immunol. 1993;24:1–12.
4. Zhang J, Patel G. Partner change and perinatal outcomes: a sys-
and accommodation of the genetically foreign fetus (toler-
tematic review. Paediatr Perinat Epidemiol. 2007;21(Suppl 1):
ization), and innate immunity, with vascular inflammation 46–57.
secondary to the stresses imposed by normal and abnor- 5. Basso O, Christensen K, Olsen J. Higher risk of pre-eclamp-
mal placentas. There is evidence that both contribute to the sia after change of partner. An effect of longer interpregnancy
pathogenesis of preeclampsia. Innate immunity has no mem- intervals? Epidemiology. 2001;12:624–629.
ory and cannot therefore explain the first-pregnancy prepon- 6. Skjaerven R, Wilcox AJ, Lie RT. The interval between
derance and possible partner specificity of preeclampsia. pregnancies and the risk of preeclampsia. N Engl J Med.
These features imply maternal tolerization to fetal (paternal) 2002;346:33–38.
antigens and suggest the existence of fetus-specific maternal 7. Zhang J. Partner change, birth interval and risk of pre-
T regulatory cells, which persist after pregnancy as memory eclampsia: a paradoxical triangle. Paediatr Perinat Epidemiol.
cells (adaptive immunity). These have been demonstrated in 2007;21(Suppl 1):31–35.
8. Saito S, Sakai M, Sasaki Y, Nakashima A, Shiozaki A.
murine but not yet in human pregnancies.
Inadequate tolerance induction may induce pre-eclampsia.
To accommodate all immune aspects into a coher- J Reprod Immunol. 2007;76:30–39.
ent model of preeclampsia, we have extended the classi- 9. Robillard PY, Hulsey TC. Association of pregnancy-induced-
cal two-stage model of preeclampsia to include four stages hypertension, pre-eclampsia, and eclampsia with duration
(Fig. 8.4). Before pregnancy there may be poor tolerization of sexual cohabitation before conception. Lancet. 1996;347
to paternal antigens, due to limited exposure to seminal anti- 619–619.
gens (Stage 1). At the end of pregnancy uteroplacental circu- 10. Robillard PY, Dekker G, Chaouat G, Hulsey TC. Etiology of
lation may be impaired by acute atherosis (Stage 4). Immune preeclampsia: maternal vascular predisposition and couple
recognition of the foreign fetus occurs at maternal-fetal disease–mutual exclusion or complementarity? J Reprod
Interface 1. Dysregulated recognition leads to poor placen- Immunol. 2007;76:1–7.
tation (new Stage 2) and abnormal uteroplacental perfusion. 11. Dekker G. The partner’s role in the etiology of preeclampsia.
J Reprod Immunol. 2002;57:203–215.
Normal pregnancy imposes substantial systemic inflam-
12. Kho EM, McCowan LM, North RA. Duration of sexual
matory stress on all pregnant women in the second half of relationship and its effect on preeclampsia and small for
pregnancy, which is associated with a Th2 immune bias. In gestational age perinatal outcome. J Reprod Immunol.
preeclampsia the vascular inflammatory response is excessive 2009;82:66–73.
with a Th1/Th17 bias. Many features that are considered to be 13. Matzinger P. The danger model: a renewed sense of self.
physiological responses to pregnancy are best considered to Science. 2002;296:301–305.
be part of an acute-phase inflammatory response. Angiogenic 14. Gordon S. Pattern recognition receptors: doubling up for the
imbalance, created by factors released from the oxidatively innate immune response. Cell. 2002;111:927–930.
stressed placenta, adds to this burden. Th17 cells promote auto- 15. Harding HP, Zhang Y, Zeng H, et  al. An integrated stress
immunity and may predispose to the generation of anti-ATR1 response regulates amino acid metabolism and resistance to
autoantibodies, which by their agonistic activity are strongly oxidative stress. Mol Cell. 2003;1:619–633.
16. Ljunggren HG, Karre K. In search of the missing self:
proinflammatory and exacerbate vascular oxidative stress.
MHC molecules and NK recognition. Immunol Today.
In Stage 3, oxidative and inflammatory stresses coalesce 1990;11:237–244.
in a vascular inflammatory response. The many conse- 17. Hunt JS. Stranger in a strange land. Immunol Rev.
quences affect numerous circulating components of the 2006;213:36–47.
inflammatory network and cause metabolic shifts including 18. Apps R, Gardner L, Moffett A. A critical look at HLA-G.
insulin resistance and dyslipidemia (Fig. 8.6). At the end of Trends Immunol. 2008;29:313–321.
pregnancy the uteroplacental circulation may be impaired 19. Redman CW, Sargent IL. Placental stress and pre-eclampsia:
by acute atherosis (Stage 4), which may result from local- a revised view. Placenta. 2009;30(Suppl A):S38–S42.
ized arterial inflammation (adaptive immunity) as in athero- 20. Sargent IL, Borzychowski AM, Redman CW. NK cells and
sclerosis or even graft vascular disease (innate immunity). human pregnancy – an inflammatory view. Trends Immunol.
2006;27:399–404.
21. Redman CW. Current topic: pre-eclampsia and the placenta.
Placenta. 1991;12:301–308.
References
22. Hanna J, Goldman-Wohl D, Hamani Y, et  al. Decidual NK
1. Billington WD. The immunological problem of pregnancy: 50 cells regulate key developmental processes at the human fetal-
years with the hope of progress. A tribute to Peter Medawar. J maternal interface. Nat Med. 2006;2:1065–1074.
Reprod Immunol. 2003;60:1–11. 23. Clements CS, Kjer-Nielsen L, McCluskey J, Rossjohn J.
2. Feeney JG, Scott JS. Pre-eclampsia and changed paternity. Eur Structural studies on HLA-G: implications for ligand and
J Obstet Gynecol Reprod Biol. 1980;11:35–38. receptor binding. Hum Immunol. 2007;68:220–226.
176 Chesley’s Hypertensive Disorders in Pregnancy

24. Parham P. MHC class I molecules and KIRs in human history, 44. Trayhurn P, Wood IS. Adipokines: inflammation and
health and survival. Nat Rev Immunol. 2005;5:201–214. the pleiotropic role of white adipose tissue. Br J Nutr.
25. Chazara O, Xiong S, Moffett A. Maternal KIR and fetal HLA- 2004;92:347–355.
C: a fine balance. J Leukoc Biol. 2011;90:703–716. 45. Ribatti D, Conconi MT, Nussdorfer GG. Nonclassic endog-
26. Rajagopalan S, Long EO. Understanding how combina- enous novel regulators of angiogenesis. Pharmacol Rev.
tions of HLA and KIR genes influence disease. J Exp Med. 2007;59:185–205.
2005;201:1025–1029. 46. Suzuki Y, Ruiz-Ortega M, Lorenzo O, Ruperez M, Esteban V,
27. Moffett A, Hiby SE. How Does the maternal immune system Egido J. Inflammation and angiotensin II. Int J Biochem Cell
contribute to the development of pre-eclampsia? Placenta. Biol. 2003;35:881–900.
2007;28(Suppl A):S51–S56. 47. Zhao Q, Ishibashi M, Hiasa K, Tan C, Takeshita A, Egashira
28. Wilczyński JR. Immunological analogy between allograft K. Essential role of vascular endothelial growth factor in
rejection, recurrent abortion and pre-eclampsia – the same angiotensin II-induced vascular inflammation and remodel-
basic mechanism? Hum Immunol. 2006;67:492–511. ling. Hypertension. 2004;44:264–270.
29. Hiby SE, Apps R, Sharkey AM, et  al. Maternal activating 48. Dandona P, Aljada A, Chaudhuri A, Mohanty P, Rajesh G.
KIRs protect against human reproductive failure mediated by A novel view of metabolic syndrome. Metab Syndr Relat
fetal HLA-C2. J Clin Invest. 2010;120:4102–4110. Disord. 2004;2:2–8.
30. van Bergen J, Koning F. The tortoise and the hare: slowly 49. Sethi JK, Hotamisligil GS. The role of TNF alpha in adipo-
evolving T-cell responses take hastily evolving KIR. cyte metabolism. Semin Cell Dev Biol. 1999;10:19–29.
Immunology. 2010;131:301–309. 50. Harris HW, Gosnell JE, Kumwenda ZL. The lipemia of sep-
31. Tilburgs T, Roelen DL, van der Mast BJ, et  al. Differential sis: triglyceride-rich lipoproteins as agents of innate immu-
distribution of CD4(+)CD25(bright) and CD8(+)CD28(−) nity. J Endotoxin Res. 2001;6:421–430.
T-cells in decidua and maternal blood during human preg- 51. Matarese G, Moschos S, Mantzoros CS. Leptin in immunol-
nancy. Placenta. 2006;27(Suppl A):S47–S53. ogy. J Immunol. 2005;174 3137–3131.
32. Redman CW, Sacks GP, Sargent IL. Preeclampsia: an exces- 52. Isomaa B. A major health hazard: the metabolic syndrome.
sive maternal inflammatory response to pregnancy. Am J Life Sci. 2003;73:2395–2411.
Obstet Gynecol. 1999;180:499–506. 53. Yoo JY, Desiderio S. Innate and acquired immunity intersect
33. Redman CW, Sargent IL, Staff AC. Making sense of pre- in a global view of the acute-phase response. Proc Natl Acad
eclampsia: two placental causes of preeclampsia? Placenta. Sci U S A. 2003;100:1157–1162.
2014;35(Suppl):S20–S25. 54. Hansson GK, Libby P, Schönbeck U, Yan ZQ. Innate and
34. Roberts JM, Taylor RN, Musci TJ, Rodgers GM, Hubel CA, adaptive immunity in the pathogenesis of atherosclerosis.
McLaughlin MK. Preeclampsia: an endothelial cell disorder. Circ Res. 2002;91:281–291.
Am J Obstet Gynecol. 1989;161:1200–1204. 55. Hotamisligil GS. Inflammation and metabolic disorders.
35. Stokes KY, Granger DN. The microcirculation: a motor Nature. 2006;444:860–867.
for the systemic inflammatory response and large ves- 56. Gabay C, Kushner I. Acute-phase proteins and other systemic
sel disease induced by hypercholesterolaemia? J Physiol. responses to inflammation. New Engl J Med. 1999;340:448–454.
2005;562:647–653. 57. Pepys MB, Hirschfield GM. C-reactive protein: a critical
36. Rath E, Haller D. Inflammation and cellular stress: a mech- update. J Clin Invest. 2003;111:1805–1812.
anistic link between immune-mediated and metabolically 58. Willis C, Morris JM, Danis V, Gallery ED. Cytokine
driven pathologies. Eur J Nutr. 2011;50(4):219–233. production by peripheral blood monocytes during the normal
37. Noonan DM, De Lerma Barbaro A, Vannini N, Mortara L, human ovulatory menstrual cycle. Hum Reprod. 2003;18:
Albini A. Inflammation, inflammatory cells and angiogen- 1173–1178.
esis: decisions and indecisions. Cancer Metastasis Rev. 59. Studd JW, Blainey JD, Bailey DE. Serum protein changes in
2008;27:31–40. the pre-eclampsia-eclampsia syndrome. J Obstet Gynaecol Br
38. Hensley K, Robinson KA, Gabbita SP, Salsman S, Floyd RA. Commonw. 1970;77:796–801.
Reactive oxygen species, cell signaling, and cell injury. Free 60. Gatti L, Tenconi PM, Guarneri D, et al. Hemostatic parameters
Radic Biol Med. 2000;28:1456–1462. and platelet activation by flow-cytometry in normal pregnancy:
39. Bastard JP, Maachi M, Lagathu C, et  al. Recent advances in a longitudinal study. Int J Clin Lab Res. 1994;24:217–219.
the relationship between obesity, inflammation, and insulin 61. Redman CW, Sargent IL. Pre-eclampsia and the systemic
resistance. Eur Cytokine Netw. 2006;17:4–12. inflammatory response. In: Belfort M, Lyall F, eds. Pre-
40. Ahn KS, Aggarwal BB. Transcription factor NF-kappaB: eclampsia – Aetiology and Clinical Practice. Cambridge:
a sensor for smoke and stress signals. Ann N Y Acad Sci. Cambridge University Press; 2004:103–120.
2005;1056:218–233. 62. Sacks GP, Seyani L, Lavery S, Trew G. Maternal C-reactive
41. Dodson M, Darley-Usmar V, Zhang J. Cellular metabolic and protein levels are raised at 4 weeks gestation. Hum Reprod.
autophagic pathways: traffic control by redox signalling. Free 2004;19:1025–1030.
Radic Biol Med. 2013;63:207–221. 63. Smarason AK, Gunnarsson A, Alfredsson JH, Valdimarsson
42. Gregor MF, Hotamisligil GS. Thematic review series: adipo- H. Monocytosis and monocytic infiltration of decidua in early
cyte biology adipocyte stress: the endoplasmic reticulum and pregnancy. J Clin Lab Immunol. 1986;21:1–5.
metabolic disease. J Lipid Res. 2007;48:1905–1914. 64. Sacks GP, Studena K, Sargent K, Redman CW. Normal preg-
43. Hotamisligil GS. Endoplasmic reticulum stress and the nancy and preeclampsia both produce inflammatory changes
inflammatory basis of metabolic disease. Cell. 2010;140: in peripheral blood leukocytes akin to those of sepsis. Am J
900–917. Obstet Gynecol. 1998;179:80–86.
Chapter 8 · Immunology of Normal Pregnancy and Preeclampsia 177

65. Vince GS, Starkey PM, Austgulen R, Kwiatkowski D, 84. Marzi M, Vigano A, Trabattoni D, et  al. Characterization
Redman CW. Interleukin-6, tumour necrosis factor and sol- of type 1 and type 2 cytokine production profile in physi-
uble tumour necrosis factor receptors in women with pre- ologic and pathologic human pregnancy. Clin Exp Immunol.
eclampsia. Br J Obstet Gynaecol. 1995;102:20–25. 1996;106:127–133.
66. Pitkin RM, Witte DL. Platelet and leukocyte counts in preg- 85. Wilczynski JR, Tchorzewski H, Banasik M, et  al.
nancy. JAMA. 1979;242:2696–2698. Lymphocyte subset distribution and cytokine secretion in
67. Richani K, Soto E, Romero R, et  al. Normal pregnancy is third trimester decidua in normal pregnancy and preeclamp-
characterized by systemic activation of the complement sys- sia. Eur J Obstet Gynecol Reprod Biol. 2003;109:8–15.
tem. J Matern Fetal Neonatal Med. 2005;17:239–245. 86. Borzychowski AM, Croy BA, Chan WL, Redman CW,
68. Chabloz P, Reber G, Boehlen F, Hohlfeld P, de Moerloose P. Sargent IL. Changes in systemic type 1 and type 2 immunity
TAFI antigen and D-dimer levels during normal pregnancy in normal pregnancy and pre-eclampsia may be mediated by
and at delivery. Br J Haematol. 2001;115:150–152. natural killer cells. Eur J Immunol. 2005;35:3054–3063.
69. Janes SL, Goodall AH. Flow cytometric detection of circulat- 87. Sacks GP, Redman CW, Sargent IL. Monocytes are primed to
ing activated platelets and platelet hyper-responsiveness in pre- express the Th1 type cytokine IL-12 in normal human preg-
eclampsia and pregnancy. Clin Sci Colch. 1994;86:731–739. nancy: an intracellular flow cytometric analysis of peripheral
70. Sørensen JD, Secher NJ, Jespersen J. Perturbed (proco- blood mononuclear cells. Clin Exp Immunol. 2003;131:490–497.
agulant) endothelium and deviations within the fibrinolytic 88. Corthay A. How do regulatory T cells work? Scand J
system during the third trimester of normal pregnancy A pos- Immunol. 2009;70:326–336.
sible link to placental function. Acta Obstet Gynecol Scand. 89. Sakaguchi S, Yamaguchi T, Nomura T, Ono M. Regulatory T
1995;74:257–261. cells and immune tolerance. Cell. 2008;133:775–787.
71. Belo L, Caslake M, Santos-Silva A, et al. LDL size, total anti- 90. Aluvihare VR, Kallikourdis M, Betz AG. Regulatory T
oxidant status and oxidised LDL in normal human pregnancy: cells mediate maternal tolerance to the fetus. Nat Immunol.
a longitudinal study. Atherosclerosis. 2004;177:391–399. 2004;5:266–271.
72. Little RE, Gladen BC. Levels of lipid peroxides in uncompli- 91. Arruvito L, Sanz M, Banham AH, Fainboim L. Expansion
cated pregnancy: a review of the literature. Reprod Toxicol. of CD4 + CD25 + and FOXP3+ regulatory T cells during
1999;13:347–352. the follicular phase of the menstrual cycle: implications for
73. Victor VM, Rocha M, Esplugues JV, De la Fuente M. Role of human reproduction. J Immunol. 2007;178:2572–2578.
free radicals in sepsis: antioxidant therapy. Curr Pharm Des. 92. Somerset DA, Zheng Y, Kilby MD, Sansom DM, Drayson
2005;11:3141–3158. MT. Normal human pregnancy is associated with an eleva-
74. Rius J, Guma M, Schachtrup C, et al. NF-kappaB links innate tion in the immune suppressive CD25 CD4 regulatory T-cell
immunity to the hypoxic response through transcriptional reg- subset. Immunology. 2004;112:38–43.
ulation of HIF-1alpha. Nature. 2008;453:807–811. 93. Mjösberg J, Berg G, Jenmalm MC, Ernerudh J. FOXP3+
75. Peritt D, Robertson S, Gri G, Showe L, Aste-Amezaga M, regulatory T cells and T helper 1, T helper 2, and T helper
Trinchieri G. Differentiation of human NK cells into NK1 and 17 cells in human early pregnancy decidua. Biol Reprod.
NK2 subsets. J Immunol. 1998;161:5821–5824. 2010;82:698–705.
76. Redman CW, Sargent IL. Immunology of pre-eclampsia. Am J 94. Tilburgs T, Scherjon SA, van der Mast BJ, et  al. Fetal-
Reprod Immunol. 2010;63:534–543. maternal HLA-C mismatch is associated with decidual T cell
77. Terrone DA, Rinehart BK, May WL, Moore A, Magann EF, activation and induction of functional T regulatory cells.
Martin NJ. Leukocytosis is proportional to HELLP syndrome J Reprod Immunol. 2009;82:148–157.
severity: evidence for an inflammatory form of preeclampsia. 95. Kudo Y, Boyd CA, Sargent IL, Redman CW. Decreased tryp-
South Med J. 2000;93:768–771. tophan catabolism by placental indoleamine 2,3-dioxygenase
78. de Messias-Reason IJ, Aleixo V, de Freitas H, Nisihara RM, in preeclampsia. Am J Obstet Gynecol. 2003;188:719–726.
Mocelin V, Urbanetz A. Complement activation in Brazilian 96. Sargent IL. Does ‘soluble’ HLA-G really exist? Another
patients with preeclampsia. J Investig Allergol Clin Immunol. twist to the tale. Mol Hum Reprod. 2005;11:695–698.
2000;10:209–214. 97. Bettelli E, Carrier Y, Gao W, et al. Reciprocal developmental
79. Conrad KP, Miles TM, Benyo DF. Circulating levels of immu- pathways for the generation of pathogenic effector TH17 and
noreactive cytokines in women with preeclampsia. Am J regulatory T cells. Nature. 2006;441:235–238.
Reprod Immunol. 1998;40:102–111. 98. Nakashima A, Ito M, Yoneda S, Shiozaki A, Hidaka T, Saito
80. Ellis J, Wennerholm UB, Bengtsson A, et  al. Levels of S. Circulating and decidual Th17 cell levels in healthy preg-
dimethylarginines and cytokines in mild and severe pre- nancy. Am J Reprod Immunol. 2010;63:104–109.
eclampsia. Acta Obstet Gynecol Scand. 2001;80:602–608. 99. Santner-Nanan B, Peek MJ, Khanam R, et  al. Systemic
81. Basu R, Hatton RD, Weaver CT. The Th17 family: flexibility increase in the ratio between Foxp3+ and IL-17-producing
follows function. Immunol Rev. 2013;252:89–103. CD4+ T cells in healthy pregnancy but not in preeclampsia.
82. Wegmann TG, Lin H, Guilbert L, Mosmann TR. Bidirectional J Immunol. 2009;183:7023–7030.
cytokine interactions in the materal-fetal relationship: is suc- 100. Sasaki Y, Darmochwal-Kolarz D, Kludka-Sternik M, et  al.
cessful pregnancy a Th2 phenomenon? Immunol Today. The predominance of Th17 lymphocytes and decreased
1993;14:353–356. number and function of Treg cells in preeclampsia. J Reprod
83. Saito S, Sakai M, Sasaki Y, Tanebe K, Tsuda H, Michimata T. Immunol. 2012;93(2):75–81.
Quantitative analysis of peripheral blood Th0, Th1, Th2 and 101. Toldi G, Svec P, Vásárhelyi B, et  al. Decreased number of
the Th1:Th2 cell ratio during normal human pregnancy and FoxP3+ regulatory T cells in preeclampsia. Acta Obstet
preeclampsia. Clin Exp Immunol. 1999;117:550–555. Gynecol Scand. 2008;87:1229–1233.
178 Chesley’s Hypertensive Disorders in Pregnancy

102. Jonsson Y, Ruber M, Matthiesen L, et al. Cytokine mapping hypertensive pregnancies. Eur J Obstet Gynecol Reprod Biol.
of sera from women with preeclampsia and normal pregnan- 1998;81:177–184.
cies. J Reprod Immunol. 2006;70:83–89. 119. Harsem NK, Roald B, Braekke K, Staff AC. Acute athero-
103. Park PH, Huang H, McMullen MR, Mandal P, Sun L, Nagy sis in decidual tissue: not associated with systemic oxidative
LE. Suppression of lipopolysaccharide-stimulated tumor stress in preeclampsia. Placenta. 2007;28:958–964.
necrosis factor-alpha production by adiponectin is mediated 120. Stevens DU, Al-Nasiry S, Bulten J, Spaanderman ME.
by transcriptional and post-transcriptional mechanisms. J Decidual vasculopathy and adverse perinatal outcome in pre-
Biol Chem. 2008;283:26850–26858. eclamptic pregnancy. Placenta. 2013;34:805–809.
104. Long EO, Kim HS, Liu D, Peterson ME, Rajagopalan S. 121. Staff AC, Dechend R, Pijnenborg R. Learning from the
Controlling natural killer cell responses: integration of placenta. Acute atherosis and vascular remodeling in pre-
signals for activation and inhibition. Annu Rev Immunol. eclampsia – novel aspects for atherosclerosis and future car-
2013;31:227–258. diovascular health. Hypertension. 2010;56:1026–1034.
105. Rowe JH, Ertelt JM, Xin L, Way SS. Pregnancy imprints 122. Staff AC, Johnsen GM, Dechend R, Redman CW.
regulatory memory that sustains anergy to fetal antigen. Preeclampsia and uteroplacental acute atherosis:
Nature. 2012;490(7418):102–106. immune and inflammatory factors. J Reprod Immunol.
106. Li XC, Zhuo JL. Nuclear factor-kappaB as a hormonal intra- 2013;S0165-0378(13):00105–00108.
cellular signaling molecule: focus on angiotensin II-induced 123. Libby P. Inflammation in atherosclerosis. Arterioscler
cardiovascular and renal injury. Curr Opin Nephrol Thromb Vasc Biol. 2012;32:2045–2051.
Hypertens. 2008;17:37–43. 124. Tsou JK, Gower RM, Ting HJ, et  al. Spatial regulation of
107. Luft FC, Dechend R, Müller DN. Immune mechanisms inflammation by human aortic endothelial cells in a linear
in angiotensin II-induced target-organ damage. Ann Med. gradient of shear stress. Microcirculation. 2008;15:311–323.
2012;44(Suppl 1):S49–S54. 125. Mitchell RN. Graft vascular disease: immune response meets
108. Hoch NE, Guzik TJ, Chen W, et  al. Regulation of T-cell the vessel wall. Annu Rev Pathol. 2009;4:19–47.
function by endogenously produced angiotensin II. Am J 126. Nickeleit V, Vamvakas EC, Pascual M, Poletti BJ, Colvin
Physiol Regul Integr Comp Physiol. 2009;296:R208–R216. RB. The prognostic significance of specific arterial lesions
109. Crispín JC, Tsokos GC. Human TCR-alpha beta+ CD4- in acute renal allograft rejection. J Am Soc Nephrol.
CD8- T cells can derive from CD8+ T cells and dis- 1998;9(7):1301–1308.
play an inflammatory effector phenotype. J Immunol. 127. Herse F, Dechend R, Harsem NK, et al. Dysregulation of the
2009;183:4675–4681. circulating and tissue-based renin-angiotensin system in pre-
110. Merrill DC, Karoly M, Chen K, Ferrario CM, Brosnihan KB. eclampsia. Hypertension. 2007;49:604–611.
Angiotensin-(1–7) in normal and preeclamptic pregnancy. 128. Angelo LS, Kurzrock R. Vascular endothelial growth factor
Endocrine. 2002;18(3):239–245. and its relationship to inflammatory mediators. Clin Cancer
111. Wallukat G, Homuth V, Fischer T, et  al. Patients with pre- Res. 2007;13:2825–2830.
eclampsia develop agonistic antibodies against the angioten- 129. Tsao PN, Chan FT, Wei SC, et  al. Soluble vascular endo-
sin AT1 receptor. J Clin Invest. 1999;1103:945–952. thelial growth factor receptor-1 protects mice in sepsis. Crit
112. Dragun D, Philippe A, Catar R, Hegner B. Autoimmune Care Med. 2007;35:1955–1960.
mediated G-protein receptor activation in cardiovascular and 130. Ahmad S, Ahmed A. Elevated placental soluble vascular
renal pathologies. Thromb Haemost. 2009;101(4):643–648. endothelial growth factor receptor-1 inhibits angiogenesis in
113. Madhur MS, Lob HE, McCann LA, et al. Interleukin 17 pro- preeclampsia. Circ Res. 2004;95:884–891.
motes angiotensin II-induced hypertension and vascular dys- 131. Selvaraj SK, Giri RK, Perelman N, Johnson C, Malik P,
function. Hypertension. 2010;55:500–507. Kalra VK. Mechanism of monocyte activation and expres-
114. Le Bouteiller P, Pizzato N, Barakonyi A, Solier C. HLA- sion of proinflammatory cytochemokines by placenta growth
G, pre-eclampsia, immunity and vascular events. J Reprod factor. Blood. 2003;102:1515–1524.
Immunol. 2003;59:219–234. 132. Taylor RN, Varma M, Teng NNH, Roberts JM. Women with
115. Blaschitz A, Juch H, Volz A, et  al. The soluble pool of preeclampsia have higher plasma endothelin levels than
HLA-G produced by human trophoblasts does not include women with normal pregnancies. J Clin Endocrinol Metab.
detectable levels of the intron 4-containing HLA-G5 and 1990;71:1675–1677.
HLA-G6 isoforms. Mol Hum Reprod. 2005;11:699–710. 133. Bernardi F, Constantino L, Machado R, Petronilho F, Dal-
116. Fons P, Chabot S, Cartwright JE, et  al. Soluble HLA-G1 Pizzol F. Plasma nitric oxide, endothelin-1, arginase and
inhibits angiogenesis through an apoptotic pathway and by superoxide dismutase in pre-eclamptic women. J Obstet
direct binding to CD160 receptor expressed by endothelial Gynaecol Res. 2008;34:957–963.
cells. Blood. 2006;108:2608–2615. 134. Kleinz MJ, Davenport AP. Emerging roles of apelin in biol-
117. Hackmon R, Koifman A, Hyodo H, Glickman H, Sheiner ogy and medicine. Pharmacol Ther. 2005;107:198–211.
E, Geraghty DE. Reduced third-trimester levels of soluble 135. Manuelpillai U, Schneider-Kolsky M, Thirunavukarasu P,
human leukocyte antigen G protein in severe preeclampsia. Dole A, Waldron K, Wallace EM. Effect of hypoxia on pla-
Am J Obstet Gynecol. 2007;197(255):e1–e5. cental activin A, inhibin A and follistatin synthesis. Placenta.
118. Hanssens M, Pijnenborg R, Keirse MJ, Vercruysse L, 2003;24:77–83.
Verbist L, Van Assche FA. Renin-like immunoreac- 136. Jones KL, Mansell A, Patella S, et al. Activin A is a critical
tivity in uterus and placenta from normotensive and component of the inflammatory response, and its binding
Chapter 8 · Immunology of Normal Pregnancy and Preeclampsia 179

protein, follistatin, reduces mortality in endotoxemia. Proc pre-eclampsia, but not normotensive intrauterine growth
Natl Acad Sci U S A. 2007;104:16239–16244. restriction. Placenta. 2006;27:56–61.
137. Fowler PA, Evans LW, Groome NP, Templeton A, 147. Smarason AK, Sargent IL, Starkey PM, Redman CW. The
Knight PG. A longitudinal study of maternal serum effect of placental syncytiotrophoblast microvillous mem-
inhibin-A, inhibin-B, activin-A, activin-AB, pro- branes from normal and pre-eclamptic women on the
alphaC and follistatin during pregnancy. Hum Reprod. growth of endothelial cells in vitro. Br J Obstet Gynaecol.
1998;13:3530–3536. 1993;100:943–949.
138. Muttukrishna S, Knight PG, Groome NP, Redman CW, 148. Southcombe J, Tannetta D, Redman C, Sargent I. The immu-
Ledger WL. Activin A and inhibin A as possible endocrine nomodulatory role of syncytiotrophoblast microvesicles.
markers for pre-eclampsia. Lancet. 1997;349:1285–1288. PLoS One. 2011;6:e20245.
139. Perkins AV, Linton EA. Identification and isolation of corti- 149. Cester N, Staffolani R, Rabini RA, et al. Pregnancy induced
cotrophin-releasing hormone-positive cells from the human hypertension: a role for peroxidation in microvillus plasma
placenta. Placenta. 1995;16:233–243. membranes. MolCell Biochem. 1994;131:151–155.
140. Perkins AV, Linton EA, Eben F, Simpson J, Wolfe CD, 150. Simons M, Raposo G. Exosomes – vesicular carriers
Redman CW. Corticotrophin-releasing hormone and corti- for intercellular communication. Curr Opin Cell Biol.
cotrophin-releasing hormone binding protein in normal and 2009;21:575–581.
pre-eclamptic human pregnancies. Br J Obstet Gynaecol. 151. Anton L, Olarerin-George AO, Schwartz N, et  al. miR-210
1995;102:118–122. inhibits trophoblast invasion and is a serum biomarker for
141. Zavalza-Gómez AB, Anaya-Prado R, Rincón-Sánche AR, preeclampsia. Am J Pathol. 2013;18:1437–1445.
Mora-Martínez JM. Adipokines and insulin resistance during 152. Visser M, Bouter LM, McQuillan GM, Wener MH, Harris
pregnancy. Diabetes Res Clin Pract. 2008;80:8–15. TB. Elevated C-reactive protein levels in overweight and
142. Ramsay JE, Ferrell WR, Crawford L, Wallace AM, Greer IA, obese adults. JAMA. 1999;282:2131–2135.
Sattar N. Divergent metabolic and vascular phenotypes in 153. Pickup JC, Chusney GD, Thomas SM, Burt D. Plasma inter-
pre-eclampsia and intrauterine growth restriction: relevance leukin-6, tumour necrosis factor alpha and blood cytokine
of adiposity. J Hypertens. 2004;22:2177–2183. production in type 2 diabetes. Life Sci. 2000;67:291–300.
143. Berckmans RJ, Neiuwland R, Böing AN, Romijn FP, Hack 154. Lacy F, O’ Connor DT, Schmid-Schönbein GW. Plasma
CE, Sturk A. Cell-derived microvesicles circulate in healthy hydrogen peroxide production in hypertensives and normo-
humans and support low grade thrombin generation. Thromb tensive subjects at genetic risk of hypertension. J Hypertens.
Haemost. 2001;85:639–646. 1998;16:291–303.
144. Redman CW, Sargent IL. Circulating microvesicles in nor- 155. Mazar RM, Srinivas SK, Sammel MD, Andrela CM, Elovitz
mal pregnancy and pre-eclampsia. Placenta. 2008;29(Suppl MA. Metabolic score as a novel approach to assessing pre-
A):S73–S77. eclampsia risk. Am J Obstet Gynecol. 2007;197(411):e1–e5.
145. Redman CW, Sargent IL. Placental debris, oxidative stress 156. Karalis K, Sano H, Redwine J, Listwak S, Wilder RL,
and pre-eclampsia. Placenta. 2000;21:597–602. Chrousos GP. Autocrine or paracrine inflammatory actions
146. Goswami D, Tannetta DS, Magee LA, et al. Excess syncytio- of corticotropin-releasing hormone in vivo. Science.
trophoblast microvesicle shedding is a feature of early-onset 1991;254:421–423.

Das könnte Ihnen auch gefallen