You are on page 1of 7

Journal of Reproductive Immunology 89 (2011) 126–132

Contents lists available at ScienceDirect

Journal of Reproductive Immunology


journal homepage: www.elsevier.com/locate/jreprimm

Review

The etiology of preeclampsia: the role of the father


Gus Dekker a,b,∗ , Pierre Yves Robillard c , Claire Roberts a
a
Discipline of Obstetrics and Gynaecology, University of Adelaide, Adelaide, South Australia 5005, Australia
b
Lyell McEwin Hospital, Medical School North, University of Adelaide, Elizabeth Vale, South Australia, Australia
c
Department of Obstetrics, Neonatology and Gynaecology, Centre -Hospitalier- Regional Reunion, France

a r t i c l e i n f o a b s t r a c t

Article history: Preeclampsia is often considered as simply a maternal disease with variable degrees of fetal
Received 4 October 2010 involvement. More and more the unique immunogenetic maternal–paternal relationship
Received in revised form
is appreciated, and also the specific ‘genetic conflict’ that is characteristic of haemochorial
29 November 2010
placentation. From that perspective, pre-eclampsia can be seen as a disease of an individual
Accepted 20 December 2010
couple with primarily maternal and fetal manifestations. The maternal and fetal genomes
perform different roles during development. Heritable paternal, rather than maternal,
imprinting of the genome is necessary for normal trophoblast development. Large pop-
Keywords:
Pre-eclampsia ulation studies have estimated that 35% of the variance in susceptibility to preeclampsia
Paternal risk factors is attributable to maternal genetic effects; 20% to fetal genetic effects (with similar con-
tributions of both parents), 13% to the couple effect, less than 1% to the shared sibling
environment and 32% to unmeasured factors. Not one of these large population studies
focussed on the paternal contribution to preeclampsia, which is demonstrated by (1) the
effect of the length of the sexual relationship; (2) the concept of primipaternity versus
primigravidity; and (3) the existence of the so-called ‘dangerous’ father, as demonstrated
in various large population studies. It is currently unknown how the father exerts this effect.
Possible mechanisms include seminal cytokine levels and their effect on maternal immune
deviation, specific paternal HLA characteristics and specific paternal single nucleotide poly-
morphisms (SNPs), in particular in the paternally expressed genes affecting placentation.
Several large cohort studies, including the large international SCOPE consortium, have
identified paternal SNPs with strong associations with preeclampsia.
Crown Copyright © 2011 Published by Elsevier Ireland Ltd. All rights reserved.

1. Introduction ically encounter preeclampsia as a maternal disease with


variable degrees of fetal involvement, not recognizing that
The etiology of preeclampsia is often considered to be preeclampsia is actually a couple’s disease with maternal
purely maternal, i.e. maternal constitutional factors that and fetal manifestations (Dekker and Robillard, 2005). The
impair maternal cardiovascular/endothelial mechanisms unique immunogenetic maternal–paternal relationship is
normally required to cope with the specific pregnancy becoming more and more appreciated, and as such also the
demands, being primarily a generalised inflammatory specific ‘genetic conflict’ that is characteristic of haemocho-
response and a hyperdynamic circulation. Clinicians typ- rial placentation (Haig, 1993).
The aim of this paper is to provide an overview on how
and to what extent paternal/relational factors play a role in
∗ Corresponding author at: Department of Obstetrics & Gynaecology, the causation of preeclampsia. Within the context of a con-
Medical School North, Lyell McEwin Hospital, University of Adelaide, Hay- cise review this paper certainly does not pretend to provide
down Road, Elizabeth Vale, South Australia 5112, Australia.
a comprehensive overview of all ‘paternal literature’; our
Tel.: +61 8 81829306; fax: +61 8 81829337.
E-mail address: gustaaf.dekker@adelaide.edu.au (G. Dekker). aim is to provide a review that reflects the broad spectrum

0165-0378/$ – see front matter. Crown Copyright © 2011 Published by Elsevier Ireland Ltd. All rights reserved.
doi:10.1016/j.jri.2010.12.010
G. Dekker et al. / Journal of Reproductive Immunology 89 (2011) 126–132 127

of paternal factors without necessarily going into too much a. Epidemiological studies suggest that advanced paternal
detail for each paternal risk factor. age is a risk factor with the risk to the mother of devel-
The clinical findings of preeclampsia can manifest as oping preeclampsia almost doubling if she has a partner
a maternal syndrome (hypertension and proteinuria with aged >45 years (Chen et al., 2006; Harlap et al., 2002). At
or without other multisystem abnormalities) and/or as the moment, the mechanism by which increased pater-
a fetal syndrome (IUGR), reduced amniotic fluid, and nal age causes an increase in maternal risk of developing
abnormal oxygenation (Redman and Sargent, 2009). The preeclampsia is still unknown. Increased sperm DNA
syndrome “preeclampsia” probably encapsulates more damage or perhaps altered sperm exposure are hypothe-
than one disease process. Thus, near-term preeclamp- ses that need further exploration.
sia without demonstrable fetal involvement is different b. Men with a familial history of early-onset cardiovascu-
from preeclampsia associated with low birth weight and lar disease and/or hypertension significantly (odds ratio
preterm delivery, and preeclampsia in nulliparous women >3) increase the risk of preeclampsia in their partners
may be different from that in women with pre-existing (Rigó et al., 2006; Dekker personal observation; SCOPE
vascular disease, multifetal gestation, diabetes mellitus, or unpublished data; see also Section 6).
previous preeclampsia (Sibai et al., 2005). The two-stage c. Paternal obesity represents an independent risk factor
model of preeclampsia envisages (Redman and Sargent, for pregnancies resulting in small-for – gestational age
2009) that preeclampsia arises in various ways includ- infants; to our knowledge no data are yet available with
ing from placental ischaemia–reperfusion injury secondary regard to the risk of; preeclampsia (McCowan et al.,
to deficient placentation. Whereas some consider abnor- 2010). Paternal obesity might not only reflect the cou-
mal placentation to be the start and invariable cause of ple’s lifestyle and dietary choices, but almost certainly
preeclampsia, it is much more likely that it is a com- also relates to the specific paternal genetic make-up (see
pletely separate, but predisposing condition. Some, but also Section 6).
certainly not all, cases of preeclampsia are associated with
poor placentation. Poor placentation defines the first stage,
Preeclampsia is a human disease. The case report on
which would appear to have a different origin; first-stage
‘eclamptic gorillas’ (Baird, 1981) reported on pregnant
decidual immune responses account for risk factors dis-
gorillas with seizures – which could have been many things
cussed later in this review like the primipaternity and
(in particular just straightforward epilepsy), but there was
possible partner specificity of preeclampsia (Redman and
no support for genuine preeclampsia/eclampsia. Why is
Sargent, 2009; Sibai et al., 2005). A systemic inflammatory
preeclampsia so uniquely a human disease? There are
response represents the second stage. Although normal
many hypotheses, but an attractive hypothesis for fur-
pregnancy stimulates a systemic inflammatory response, in
ther research would be that women, in contrast to other
a completely normal woman this is not intense enough to
mammals cannot identify a ‘suitable match’. Could there
generate the signs of preeclampsia. To do that requires the
be a specifically human lack of pheromone recognition –
abnormal stimulus from an oxidatively stressed placenta
after all pheromones certainly have an association with the
(“placental preeclampsia”). In a woman with a chronic
MHC (Scott, 2003). Although most data are by default ani-
systemic inflammatory response associated with condi-
mal studies (Grob et al., 1998), there are certainly some
tions, such as chronic hypertension, diabetes, or obesity,
human data suggesting that human partner selection on
the starting point is abnormal enough that even a nor-
an olfactory basis would favour HLA (MHC) heterozygos-
mal placenta can stimulate a systemic response of such an
ity (Wedekind and Füri, 1997; see also Section 3), while de
intensity as to give the signs of preeclampsia (“maternal
Luca Brunori et al. (2003) have described a statistically sig-
preeclampsia”; Redman and Sargent, 2009). In a previous
nificant increase in HLA-DR homozygosity and a reduced
review, Robillard et al. (2007) stressed that the presence
antigenic variety in ‘preeclamptic couples’ versus control
of these two major subtypes of preeclampsia probably
couples. However, for practical purposes these potential
underlies the many major disagreements in the literature
feminine recognition strategies might not be very practical
regarding cause, risk factors and long-term prognosis of
in daily human life.
preeclampsia. Although many if not virtually all articles
reviewed in this paper do not specifically define which
subtype of preeclampsia was addressed in their respective 3. The partner’s HLA and parental ethnic
studies, the focus of this paper on paternal risk factors is by disconcordance
default on that type of preeclampsia that is associated with
defective placentation. The invasion of trophoblasts into the decidua and
myometrium appears to be primarily controlled by
immune mechanisms. The syncytiotrophoblast does not
2. The role of the father express class I HLA mRNA. While all of the classical class
I HLA antigens are absent apart from HLA-C, the invad-
The concept of the dangerous partner is certainly not ing cytotrophoblasts express the non-classical HLA-G and
new. Twenty years ago Astin et al. (1981) published a HLA-E. Initially it was thought that the presence of HLA-G,
paper in The Lancet entitled “Preeclampsia/eclampsia: a being monomorphic, played an important role in the affer-
fatal father factor”. It might be difficult for a woman to iden- ent arm of maternal tolerance to the fetus. HLA-G allows
tify her ideal partner, but some potential paternal factors trophoblasts to evade detection as foreign and prevents NK
are at least in theory relatively easily recognisable: cell mediated cytotoxicity.
128 G. Dekker et al. / Journal of Reproductive Immunology 89 (2011) 126–132

We now know that HLA-G expressed on the invad- have now demonstrated that specific paternal (fetal) HLA-
ing cytotrophoblast plays an even more important role G polymorphisms or deletions (Tan et al., 2008, Larsen et al.,
in the vascular adaptations in the placental bed that 2010) are risk factors for developing preeclampsia.
are essential to pregnancy success (Moffett-King, 2002).
Since T-cells were thought to be the unique immune 4. Sperm exposure
cells required for adaptive immune responses, the absence
of major T-cell interaction in preeclampsia appeared to Most, but not all, epidemiological studies demonstrate
negate the Immune Maladaptation Hypothesis (Dekker and that regular sexual intercourse over an extended period
Sibai, 1998). This concept was radically changed by the reduces the risk of preeclampsia. Marti and Herrmann
realisation of the major role played by the decidual NK (1977) were the first to suggest that repeated prior expo-
cells. The predominant population of lymphoid cells in sure to semen from the biological father of the baby reduces
the decidua consists of NK cells while T and B cells are the risk of preeclampsia, followed by the landmark study
rare. NK cells express inhibitory and activatory killer-cell of Robillard et al. (1994). Although most subsequent retro-
immunoglobulin-like receptors (KIR) capable of recognis- spective studies concurred with this finding (Dekker and
ing HLA-class I molecules. Uterine NK cells influence both Robillard, 2007), a 2004 prospective study in women of
trophoblast invasion and the vascular transformation of the mixed parity found no association between duration of
maternal placental bed by producing a series of cytokines sexual relationship with the biological father and the risk
involved in angiogenesis and vascular stability, in particu- of preeclampsia (Ness et al., 2004). However, in a recent
lar VEGF, PlGF and Ang-2 (Lash et al., 2006). Moffett-King large prospective cohort study (Kho et al., 2009), 2507
(2002) changed our understanding of the importance of nulliparous women with singleton pregnancies were inter-
the HLA-C–NK cell receptor interaction. HLA-C loci are viewed at 15 weeks’ gestation about the duration of their
dimorphic for residues 77–80 and these two HLA-C groups sexual relationship with the biological father. This study
interact with different NK cell receptors. There is great clearly showed that a short duration of sexual relation-
diversity of NK KIR haplotypes in humans with variations in ship was more common in women with preeclampsia
the number of genes, as well as polymorphisms at individ- compared with uncomplicated pregnancies (≤6 months
ual loci. All women express KIR on decidual NK cells for both 14.5% versus 6.9%, adjusted odds ratio [adjOR] 1.88, 95% CI
groups of HLA-C alleles and because HLA-C is polymor- 1.05–3.36; ≤3 months 6.9% versus 2.5%, adjOR 2.32, 95% CI
phic, each pregnancy will involve different combinations 1.03–5.25; first intercourse 1.5% versus 0.5%, adjOR 5.75,
of paternally derived fetal HLA-C and maternal KIRs. 95% CI 1.13–29.3). Although the total number of semen
Therefore each pregnancy is associated with a specific exposures was lower in women who delivered small-for-
immune interaction involving maternal NK cells inter- gestational-age (SGA) babies, SGA was not associated with
acting with ‘paternal’ HLA. Mothers lacking most or all a shorter duration of the sexual relationship. On post hoc
activating KIRs (AA genotype) when the fetus has HLA-C analysis, the subgroup of mothers with SGA fetuses and
belonging to the HLA-C2 group, are at substantial risk of abnormal uterine artery Doppler at 20 weeks was more
preeclampsia (Hiby et al., 2004). An important component likely to have had a short sexual relationship compared
of the Hiby et al. (2004) paper was the finding that dif- with controls (≤6 months adjOR 2.33, 95% CI 1.09–4.98;
ferent human populations have a reciprocal relationship ≤3 months adjOR 3.22, 95% CI 1.18–8.79; first intercourse
between KIR AA frequency and HLA-C2 frequency. Thereby, adjOR 8.02, 95% CI 1.58–40.7). The authors concluded that
this very attractive hypothesis could provide an immuno- compared with uncomplicated pregnancies, a short dura-
logical explanation for the various studies on parental tion of sexual relationship is more common in women who
ethnic disconcordance or racial dissimilarity as a risk fac- develop preeclampsia and women with abnormal uter-
tor for preeclampsia. Indeed, Alderman et al. (1986), based ine artery Doppler waveforms who deliver a SGA baby.
on a large Washington cohort, reported an almost dou- Interestingly, the results of Kho et al. (2009) indicate that
bling of the risk of preeclampsia associated with racial there is no relation between the length of the sexual rela-
dissimilarity – in this particular cohort primarily Cau- tionship and gestational hypertension. However, a recent
casian versus African-American. More recently, Caughey paper from Nigeria by Olayemi et al. (2010) reports a
et al. (2005), although confirming the risk associated with 4% drop in the risk of developing hypertension for every
parental ethnic disconcordance, came to much lower risk month increase in sexual cohabitation, but this did not
estimates. According to the Hiby hypothesis the incidence apply to preeclampsia. It should be noted that this Nige-
of preeclampsia among couples consisting of Japanese rian study also confirmed risk reduction (hazard ratio 0.7;
women and Caucasian men should be higher than that 95% CI 0.55–0.93) associated with same-paternity abor-
among couples consisting of Japanese women and Japanese tion.
men. However, Saito et al. (2006) investigated the inci- The ‘protective’ effect of a more lengthy sexual rela-
dence of preeclampsia among 324 couples consisting of tionship is explained by maternal mucosal tolerance to
Japanese women and Caucasian men and found it to be paternal antigens (Robertson et al., 2002; Peters et al.,
similar to that in couples consisting of Japanese women 2004). Deposition of semen in the female genital tract
and Japanese men. The authors concluded, therefore, that elicits a cascade of cellular and molecular events that
their data do not support those of Hiby et al. (2004). resembles a classic inflammatory response (Sharkey et al.,
Although HLA-G was initially seen as essentially 2007). Seminal vesicle-derived transforming growth fac-
monomorphic, more recent data have shown that this tor ␤ (TGF␤) is an important contributing factor. Seminal
might not be the case (Walpole et al., 2010). Various studies fluid TGF␤ is present both as a soluble form at five
G. Dekker et al. / Journal of Reproductive Immunology 89 (2011) 126–132 129

times the concentration of that in blood (Nocera and is the study by Lie et al. (1998). These authors have shown
Chu, 1995), as well as bound to sperm (Chu et al., 1996). that men who fathered one preeclamptic pregnancy were
TGF␤ elicits strong type 2 and Th3 immune responses almost twice as likely to father a preeclamptic pregnancy
towards antigens present in semen (Robertson et al., with a different woman. While Esplin et al. (2001) showed
2002). The antigenic stimulus is likely to be conveyed that if the male partner was born of a pregnancy compli-
by sperm, since the risk of preeclampsia is three times cated by preeclampsia, the risk of his partner developing
higher in women conceiving via intracytoplasmic sperm preeclampsia would more than double. Emerging evidence
injection (ICSI) with surgically obtained sperm (from men dictates that we should no longer neglect the role that the
with complete azoospermia) than in those with stan- male partner might play in reproductive success. Indeed,
dard in vitro fertilisation and ICSI using sperm obtained male fetuses are less likely to survive at all time post-
by masturbation (Wang et al., 2002). Repeated inter- conception than females, and in particular male fetuses
course with sustained antigen exposure in the appropriate are more likely to be born preterm (Vatten and Skjaerven,
cytokine environment mediated by TGF␤ is now thought 2004). Some of this biology appears to relate to gender-
to be essential in this partner-specific mucosal tolerance specific ways in which the fetus responds to stress (Clifton,
(Robertson et al., 2002). By initiating a type 2 or T regulatory 2010).
cell-dominated immune response towards paternal anti- In addition to the aforementioned strict Mendelian
gens (Robertson et al., 2009), seminal TGF␤ may inhibit the laws, we also have to consider the potential involvement
induction of type 1 responses against the semi-allogeneic impact of imprinted genes, and of genes that could change
conceptus that are thought to be associated with poor part of his physiology relevant to reproduction. A few
placental development and restricted fetal growth. Fur- examples of these three ‘genetic’ possibilities are:
thermore, T helper type 2 (Th2) and Th3 lymphocytes
interact with both placental trophoblasts and the mater-
nal decidual vasculature to facilitate their remodelling • Dangerous paternal genes following Mendelian law: within
(Leonard et al., 2006). Impaired spiral arterial remodelling the context of this review it is not possible to report
reduces utero-placental perfusion and thereby predisposes in detail on this rapidly evolving field. A few exam-
to preeclampsia and SGA. One of the important novel find- ples will be highlighted to illustrate the principle. The
ings of the aforementioned Kho et al. (2009) study is that aforementioned studies on paternal family history of car-
longer sexual relationships also provide partial protection diovascular disease and hypertension (Rigó et al., 2006)
against SGA associated with abnormal uterine Doppler flow are nothing less than an epidemiological reflection of
patterns. a whole set of susceptibility genes passed through to
the feto-placental unit via the father. Paternal (or fetal
5. Primipaternity autosomal) genetic thrombophilias have been linked to
a range of adverse pregnancy outcomes ranging from
Within the context of this review we will not reiter- stillbirth, to intra-uterine growth restriction (IUGR) and
ate our concerns regarding the ‘birth-interval hypothesis’ preeclampsia (de Galan-Roosen et al., 2005; Gibson et al.,
as opposed to the primipaternity hypothesis, since this 2006). Andraweera et al. (2010) published data showing
has been the subject of an earlier specific review (Dekker almost a doubling of the risk associated with paternal
and Robillard, 2003). However, it should be noted that the SNPs involving VEGF and PlGF, while Zusterzeel et al.
primipaternity hypothesis does not reject the possibility (2002) showed that the polymorphism in the glutathione
of a relevant biological effect of long intervals between S-transferase P1 gene, a major biotransformation enzyme
births. The key point, however, is that the vast major- in placenta and decidua associated with lower enzyme
ity of studies examining the effect of paternity change, detoxification capacity, is associated with preeclampsia
including the recent large study from Nigeria (Olayemi irrespective of whether it was of maternal or paternal
et al., 2010), confirms the increased risk associated with a origin. The potential involvement of some other paternal
change in paternity; an effect in those studies that cannot SNPs will be further discussed (see below).
be explained by the very modest increase in risk associated • Dangerous paternal genes and imprinting: epigenetic
with very long birth intervals (Skjaerven et al., 2002). From features, i.e. genomic imprinting are almost certainly
a primary preventative perspective the message is clear-for involved in the pathogenesis of preeclampsia (Cross,
the best chance on having an uncomplicated second preg- 2003). Direct proof of the role of imprinting was pub-
nancy, if your first pregnancy was uncomplicated, have it lished by Oudejans et al. (2004) who confirmed the
with the same man. susceptibility locus on chromosome 10q22.1, previously
identified by Lachmeijer et al. (2001). Haplotype analysis
6. Dangerous paternal genes showed a parent-of-origin effect: maximal allele sharing
in the affected siblings was found for maternally derived
Large, mostly Scandinavian, studies have shown that for alleles in all families, but not for the paternally derived
preeclampsia the genetic effects account for about 31% of alleles. Single nucleotide polymorphisms in the IGF2
the variation in the propensity to develop preeclampsia gene, which encodes the paternally expressed insulin-
whereby the environment accounts for 63% (Nilsson et al., like growth factor II (IGF-II), an important growth factor
2004). Although we are all aware of the Mendelian laws, it is in trophoblast invasion and placental function (Roberts,
interesting to note that in these large epidemiological stud- 2010), are associated with a number of adverse outcomes
ies ‘men’ appear to be a ‘neglected species’. An exception of pregnancy.
130 G. Dekker et al. / Journal of Reproductive Immunology 89 (2011) 126–132

• Paternal genes changing aspects of male reproductive phys- There are several possible pathways by which certain
iology: nothing has been published on this aspect of viruses, and in particular CMV, would lead to adverse preg-
reproductive physiology as far as we are aware. However, nancy outcome and more specifically preeclampsia:
one would expect paternal SNPs to influence seminal
fluid cytokine patterns and as such adversely affect (1) Human CMV has been shown to impair clinical aspects
the required mucosal immunological adaptive changes of cytotrophoblast function (Fisher et al., 2000) such
(Robertson et al., 2002) and/or the paternal SNPs might as decreased ␣1␤1 integrin expression and decreased
influence his immune defence capacity to respond appro- trophoblast invasion, and interestingly also decreased
priately to genital tract infections (see below). HLA-G expression.
(2) CMV can persist in a latent phase in endothelial cells
7. The father as source of infection and may therefore cause chronic endothelial cell dys-
function (Jarvis and Nelson, 2002).
Over the past decade, researchers have started to look
for evidence of an infectious process underlying or at But what about the potential link between the father
least triggering the inflammatory pathways involved in and viral presence versus adverse pregnancy outcome?
preeclampsia (Trogstad et al., 2001; Xie et al., 2010; Conde- The connection might well be explained by the fact that
Agudelo et al., 2008). As a post hoc analysis of their the male urogenital tract is a major reservoir for CMV
cerebral palsy cohort, Gibson et al. (2008a) checked for pos- (Dejucq and Jégou, 2001) with various studies showing
itive PCR for neurotropic viruses using newborn screening CMV to be present in semen in 5–10% of men (Bezold
cards of 717 adverse pregnancy cases and 609 controls. et al., 2007; Kapranos et al., 2003). A chronic CMV pres-
For both term and preterm births, the risk of developing ence in the male genital tract could, at least in theory,
pregnancy-induced hypertensive disorders was increased easily change the cytokine levels in seminal fluid and as
in the presence of DNA from any herpesvirus (OR 5.70, 95 such adversely affect the aforementioned partner-specific
CI 1.85–17.57), and in particular with the presence of CMV mucosal tolerance (Robertson et al., 2002). As mentioned
(OR 3.89, 95% CI 1.67–9.06). earlier, the male’s genotype could further influence his
Using the same cohort, all adverse pregnancy outcome response towards the presence of infection, adding another
babies and controls were also genotyped for Mannose layer of complexity. In our opinion this opens a whole new
Binding Lectin (MBL) SNPs (Ozdemir et al., 2010). The area of exciting research, all aimed at unravelling the com-
MBL2 pathway of the complement system is activated plex genotype–phenotype interaction between a women
when MBL2 recognises and binds to carbohydrates, such and her male partner.
as mannose- and N-acetyl-glucosamine-rich oligosaccha-
rides, present on a wide range of bacteria, viruses, fungi
References
and parasites (Matsushita et al., 1998; Petersen et al., 2000).
The human MBL2 gene on chromosome 10 has 6 known Alderman, B.W., Sperling, R.S., Daling, J.R., 1986. An epidemiological study
polymorphic sites (Mullighan et al., 2000), three within the of the immunogenetic aetiology of preeclampsia. Br. Med. J. (Clin. Res.
promoter region of exon 1, and 3 within exon 1. These pro- Ed.) 292, 372–374.
Andraweera, P., Thompson, S., Zhang, V., Nowak, R., Dekker, G., Roberts, C.,
moter and structural polymorphisms are found in various
2010. Maternal, paternal and fetal single nucleotide polymorphisms in
cis combinations, resulting in haplotypes associated with vascular endothelial growth factor family genes associate with preg-
high, intermediate and low levels of MBL2 (Madsen et al., nancy complications. Am. J. Obstet. Gynecol. 201, S13 (SMFM abstract
220).
1995). Seven different haplotypes have been identified in
Astin, M., Scott, J.R., Worley, R.J., 1981. Preeclampsia/eclampsia: a fatal
a range of populations, and in order of decreasing levels of father factor. Lancet 5 (2), 533.
MBL2 are: HYPA, LYQA, LYPA, LXPA, HYPD, LYQC, and LYPB. Baird Jr., J.N., 1981. Eclampsia in a lowland gorilla. Am. J. Obstet. Gynecol.
The HYPD, LYQC and LYPB haplotypes are considered defec- 141, 345–346.
Bezold, G., Politch, J.A., Kiviat, N.B., Kuypers, J.M., Wolff, H., Anderson,
tive haplotypes because they are associated with virtually D.J., 2007. Prevalence of sexually transmissible pathogens in semen
undetectable concentrations of MBL2 (Turner and Hamvas, from asymptomatic male infertility patients with and without leuko-
2000). These so-called deficient MBL2 haplotypes LYPA or cytospermia. Fertil. Steril. 87, 1087–1097.
Caughey, A.B., Stotland, N.E., Washington, A.E., Escobar, G.J., 2005. Mater-
HYPD may be associated with an increased risk of cere- nal ethnicity, paternal ethnicity, and parental ethnic discordance:
bral palsy in the presence of exposure to viral infection and predictors of preeclampsia. Obstet. Gynecol. 106, 156–161.
may act as susceptibility factors for cerebral palsy (Gibson Chen, X.K., Wen, S.W., Smith, G., Leader, A., Sutandar, M., Yang, Q., Walker,
M., 2006. Maternal age, paternal age and new-onset hypertension in
et al., 2008b). The same group also looked at deficient MBL late pregnancy. Hypertens. Pregnancy 25, 217–227.
haplotypes, viral presence and pregnancy outcome; sig- Clifton, V.L., 2010. Review: sex and the human placenta: mediating differ-
nificant associations were found between variant MBL2 ential strategies of fetal growth and survival. Placenta 31, S33–S39.
Chu, T.M., Nocera, M.A., Flanders, K.C., Kawinski, E., 1996. Localization of
haplotypes and pregnancy-induced hypertensive disorders
seminal plasma transforming growth factor-beta1 on human sperma-
(PIHD) (LYQC <32 weeks (OR 17.89, 95% CI 2.20–139.57)). tozoa: an immunocytochemical study. Fertil. Steril. 66, 327–330.
Evidence of exposure to infection increased the effect of Conde-Agudelo, A., Villar, J., Lindheimer, M., 2008. Maternal infection
and risk of preeclampsia: systematic review and metaanalysis. Am.
these associations (PIHD OR 23.80, 95% CI 1.08–1414.76)
J. Obstet. Gynecol. 198, 7–22.
while in the absence of evidence of exposure to infection Cross, J.C., 2003. The genetics of preeclampsia: a feto-placental or maternal
demonstrated that no associations were found between problem? Clin. Genet. 64, 96–103.
deficient MBL haplotypes and PIHD (Gibson et al., 2010). de Galan-Roosen, A.E., Kuijpers, J.C., Rosendaal, F.R., Steegers, E.A., van
Beers, W.A., Ponjee, G.A., Merkus, H.M., 2005. Maternal and pater-
Of course the father directly contributes to the fetal MBL nal thrombophilia: risk factors for perinatal mortality. Br. J. Obstet.
haplotype. Gynaecol. 112, 306–311.
G. Dekker et al. / Journal of Reproductive Immunology 89 (2011) 126–132 131

Dejucq, N., Jégou, B., 2001. Viruses in the mammalian male genital tract Leonard, S., Murrant, C., Tayade, C., van den Heuvel, M., Watering, R., Croy,
and their effects on the reproductive system. Microbiol. Mol. Biol. Rev. B.A., 2006. Mechanisms regulating immune cell contributions to spiral
65, 208–231. artery modification – facts and hypotheses – a review. Placenta 27 S,
Dekker, G., Robillard, P.Y., 2003. The birth interval hypothesis – does it 40–46.
really indicate the end of the primipaternity hypothesis? J. Reprod. Lie, R.T., Rasmussen, S., Brunborg, H., Gjessing, H.K., Lie-Nielsen, E., Irgens,
Immunol. 59, 245–251. L.M., 1998. Fetal and maternal contributions to risk of preeclampsia:
Dekker, G.A., Robillard, P.Y., 2005. Preeclampsia: a couple’s disease a population based study. Br. Med. J. 316, 1343–1347.
with maternal and fetal manifestations. Curr. Pharm. Des. 11, 699– Madsen, H.O., Garred, P., Thiel, S., Kurtzhals, J.A., Lamm, L.U., Ryder,
710. L.P., Svejgaard, A., 1995. Interplay between promoter and structural
Dekker, G., Robillard, P.Y., 2007. Preeclampsia: is the immune maladapta- gene variants control basal serum level of mannan-binding protein. J.
tion hypothesis still standing? An epidemiological update. J. Reprod. Immunol. 155, 3013–3020.
Immunol. 76, 8–16. Marti, J.J., Herrmann, U., 1977. Immunogestosis: a new etiologic concept of
Dekker, G.A., Sibai, B.M., 1998. Etiology and pathogenesis of preeclamp- “essential” EPH gestosis, with special consideration of the primigravid
sia: current concepts. Am. J. Obstet. Gynecol. 179, 1359– patient: preliminary report of a clinical study. Am. J. Obstet. Gynecol.
1375. 128, 489–493.
de Luca Brunori, I., Battini, L., Simonelli, M., Brunori, E., Valentino, V., Matsushita, M., Hijikata, M., Ohta, Y., Iwata, K., Matsumoto, M., Nakao,
Curcio, M., Mariotti, M.L., Lapi, S., Genazzani, A.R., 2003. HLA-DR in K., Kanai, K., Yoshida, N., Baba, K., Mishiro, S., 1998. Hepatitis C virus
couples associated with preeclampsia: background and updating by infection and mutations of mannose-binding lectin gene MBL. Arch.
DNA sequencing. J. Reprod. Immunol. 59, 235–243. Virol 143, 645–651.
Esplin, M.S., Fausett, M.B., Fraser, A., et al., 2001. Paternal and maternal McCowan, L., North, R., Kho, E.M., Black, M., Chan, E., Dekker, G., Poston,
components of the predisposition to preeclampsia. N. Engl. J. Med. L., Taylor, R., Claire, R., 2010. Paternal contribution to small for ges-
344, 867–872. tational age babies: a multicentre prospective study. Obesity (Silver
Fisher, S., Genbacev, O., Maidji, E., Pereira, L., 2000. Human Spring), doi:10.1038/oby.2010.279.
cytomegalovirus infection of placental cytotrophoblasts in vitro Moffett-King, A., 2002. Natural killer cells and pregnancy. Nat. Rev.
and in utero: implications for transmission and pathogenesis. J. Virol. Immunol. 2, 656–663.
74, 6808–6820. Mullighan, C.G., Marshall, S.E., Welsh, K.I., 2000. Mannose binding lectin
Gibson, C.S., MacLennan, A.H., Janssen, N.G., Kist, W.J., Hague, W.M., polymorphisms are associated with early age of disease onset and
Haan, E.A., Goldwater, P.N., Priest, K., Dekker, G.A., South Australian autoimmunity in common variable immunodeficiency. Scand. J.
Cerebral Palsy Research Group, 2006. Associations between fetal Immunol. 51, 111–122.
inherited thrombophilia and adverse pregnancy outcomes. Am. J. Ness, R.B., Markovic, N., Harger, G., Day, R., 2004. Barrier methods, length of
Obstet. Gynecol. 194 (947), e1–e10. preconception intercourse, and preeclampsia. Hypertens Pregnancy
Gibson, C.S., Goldwater, P.N., MacLennan, A.H., Haan, E.A., Priest, K., 23, 227–235.
Dekker, G.A., 2008a. Fetal exposure to herpesviruses may be asso- Nilsson, E., Salonen Ros, H., Cnattingius, S., Lichtenstein, P., 2004. The
ciated with pregnancy-induced hypertensive disorders and preterm importance of genetic and environmental effects for preeclampsia and
birth in a Caucasian population. BJOG 115, 492–500. gestational hypertension: a family study. BJOG 111, 200–206.
Gibson, C.S., MacLennan, A.H., Goldwater, P.N., Haan, E.A., Priest, K., Nocera, M., Chu, T.M., 1995. Characterization of latent transforming
Dekker, G.A., 2008b. Mannose-binding lectin haplotypes may be asso- growth factor-beta from human seminal plasma. Am. J. Reprod.
ciated with cerebral palsy only after perinatal viral exposure. Am. J. Immunol. 33, 282–291.
Obstet. Gynecol. 198 (509.), e1-509.e8. Olayemi, O., Strobino, D., Aimakhu, C., Adedapo, K., Kehinde, A., Odukogbe,
Gibson, C.S., MacLennan, A.H., Haan, E.A., Priest, K., Dekker, G.A., Writ- A.T., Salako, B., 2010. Influence of duration of sexual cohabitation on
ing for the South Australian Cerebral Palsy Research Group, 2010. the risk of hypertension in nulliparous parturients in Ibadan: a cohort
Fetal MBL2 haplotypes combined with viral exposure are associated study. Aust. N. Z. J. Obstet. Gynaecol. 50, 40–44.
with adverse pregnancy outcomes. J. Matern. Fetal Neonatal Med., Oudejans, C.B., Mulders, J., Lachmeijer, A.M., Van Dijk, M., Konst, A.A.,
doi:10.3109/14767058.2010.531324. Westerman, B.A., Van Wijk, I.J., Leegwater, P.A., Kato, H.D., Matsuda,
Grob, B., Knapp, L.A., Martin, R.D., Anzenberger, G., 1998. The major T., Wake, N., Dekker, G.A., Pals, G., Ten Kate, L.P., Blankenstein, M.A.,
histocompatibility complex and mate choice: inbreeding avoidance 2004. The parent-of-origin effect of 10q22 in preeclamptic females
and selection of good genes. Exp. Clin. Immunogenet. 15, 119– coincides with two regions clustered for genes with down-regulated
129. expression in androgenetic placentas. Mol. Hum. Reprod. 10, 589–
Haig, D., 1993. Genetic conflicts in human pregnancy. Q. Rev. Biol. 68, 598.
495–532. Ozdemir, O., Dinleyici, E.C., Tekin, N., Colak, O., Aksit, M.A., 2010. Low-
Harlap, S., Paltiel, O., Deutsch, L., Knaanie, A., Masalha, S., Tiram, E., Caplan, mannose-binding lectin levels in susceptibility to neonatal sepsis
L.S., Malaspina, D., Friedlander, Y., 2002. Paternal age and preeclamp- in preterm neonates with fetal inflammatory response syndrome. J.
sia. Epidemiology 13, 660–667. Matern. Fetal Neonatal Med. Early Online, 1–5.
Hiby, S.E., Walker, J.J., O’Shaughnessy, K.M., et al., 2004. Combinations of Peters, B., Whittall, T., Babaahmady, K., Gray, K., Vaughan, R., Lehner, T.,
maternal KIR and fetal HLA-C genes influence the risk of preeclampsia 2004. Effect of heterosexual intercourse on mucosal alloimmunisation
and reproductive success. J. Exp. Med. 200, 957–965. and resistance to HIV-1 infection. Lancet 363, 518–524.
Jarvis, M.A., Nelson, J.A., 2002. Human cytomegalovirus persistence and Petersen, S.V., Thiel, S., Jensen, L., Vorup-Jensen, T., Koch, C., Jensenius, J.C.,
latency in endothelial cells and macrophages. Curr. Opin. Microbiol. 2000. Control of the classical and the MBL pathway of complement
5, 403–407. activation. Mol. Immunol. 37, 803–811.
Kapranos, N., Petrakou, E., Anastasiadou, C., Kotronias, D., 2003. Detection Redman, C.W., Sargent, I.L., 2009. Placental stress and preeclampsia: a
of herpes simplex virus, cytomegalovirus, and Epstein–Barr virus in revised view. Placenta 30, S38–42.
the semen of men attending an infertility clinic. Fertil. Steril. 79 (Suppl. Rigó Jr., J., Boze, T., Derzsy, Z., Derzbach, L., Treszl, A., Lázár, L., Sobel, G.,
3), 1566–1570. Vásárhelyi, B., 2006. Family history of early-onset cardiovascular dis-
Kho, E.M., McCowan, L.M., North, R.A., Roberts, C.T., Chan, E., Black, M.A., orders is associated with a higher risk of severe preeclampsia. Eur. J.
Taylor, R.S., Dekker, G.A., SCOPE Consortium, 2009. Duration of sexual Obstet. Gynecol. Reprod. Biol. 128, 148–151.
relationship and its effect on preeclampsia and small for gestational Roberts, C.T., 2010. IFPA award in placentology lecture: complicated
age perinatal outcome. J. Reprod. Immunol. 82, 66–73. interactions between genes and the environment in placentation,
Lachmeijer, A.M., Arngrimsson, R., Bastiaans, E.J., Frigge, M.L., Pals, G., pregnancy outcome and long term health. Placenta 31 (Suppl.),
Sigurdardottir, S., Stefansson, H., Palsson, B., Nicolae, D., Kong, A., S47–53.
Aarnoudse, J.G., Gulcher, J.R., Dekker, G.A., ten Kate, L.P., Stefansson, Robertson, S.A., Ingman, W.V., O’Leary, S., Sharkey, D.J., Tremellen, K.P.,
K., 2001. A genome-wide scan for preeclampsia in the Netherlands. 2002. Transforming growth factor beta – a mediator of immune devi-
Eur. J. Hum. Genet. 9, 758–764. ation in seminal plasma. J. Reprod. Immunol. 57, 109–128.
Larsen, M.H., Hylenius, S., Andersen, A.M., Hviid, T.V., 2010. The 3 - Robertson, S.A., Guerin, L.R., Bromfield, J.J., Branson, K.M., Ahlström, A.C.,
untranslated region of the HLA-G gene in relation to preeclampsia: Care, A.S., 2009. Seminal fluid drives expansion of the CD4 + CD25+ T
revisited. Tissue Antigens 75, 253–261. regulatory cell pool and induces tolerance to paternal alloantigens in
Lash, G.E., Schiessl, B., Kirkley, M., Innes, B.A., Cooper, A., Searle, R.F., Rob- mice. Biol. Reprod. 80, 1036–1045.
son, S.C., Bulmer, J.N., 2006. Expression of angiogenic growth factors Robillard, P.Y., Hulsey, T.C., Perianin, J., Janky, E., Miri, E.H., Papiernik, E.,
by uterine natural killer cells during early pregnancy. J. Leukoc. Biol. 1994. Association of pregnancy induced hypertension with duration
80, 572–580. of sexual cohabitation before conception. Lancet 344, 973–975.
132 G. Dekker et al. / Journal of Reproductive Immunology 89 (2011) 126–132

Robillard, P.Y., Dekker, G., Chaouat, G., Hulsey, T.C., 2007. Etiology of Turner, M.W., Hamvas, R.M., 2000. Mannose-binding lectin: structure,
preeclampsia: maternal vascular predisposition and couple disease function, genetics and disease associations. Rev. Immunogenet. 2,
– mutual exclusion or complementarity? J. Reprod. Immunol. 76, 1–7. 305–322.
Saito, S., Takeda, Y., Sakai, M., Nakabayahi, M., Hayakawa, S., 2006. The inci- Vatten, L.J., Skjaerven, R., 2004. Offspring sex and pregnancy outcome by
dence of preeclampsia among couples consisting of Japanese women length of gestation. Early Hum. Dev. 76, 47–54.
and Caucasian men. J. Reprod. Immunol. 70, 93–98. Walpole, N.G., Kjer-Nielsen, L., Kostenko, L., McCluskey, J., Brooks, A.G.,
Scott, K., 2003. Sex and the MHC. Dev. Cell 4, 290–291. Rossjohn, J., Clements, C.S., 2010. The structure and stability of the
Sharkey, D.J., Macpherson, A.M., Tremellen, K.P., Robertson, S.A., 2007. monomorphic HLA-G are influenced by the nature of the bound pep-
Seminal plasma differentially regulates inflammatory cytokine gene tide. J. Mol. Biol. 397, 467–480.
expression in human cervical and vaginal epithelial cells. Mol. Hum. Wang, J.X., Knottnerus, A.M., Schuit, G., Norman, R.J., Chan, A., Dekker, G.A.,
Reprod. 13, 491–501. 2002. Surgically obtained sperm, and risk of gestational hypertension
Sibai, B., Dekker, G., Kupferminc, M., 2005. Preeclampsia. Lancet 365, and preeclampsia. Lancet 359, 673–674.
785–799. Wedekind, C., Füri, S., 1997. Body odour preferences in men and women:
Skjaerven, R., Wilcox, A.J., Lie, R.T., 2002. The interval between pregnancies do they aim for specific MHC combinations or simply heterozygosity?
and the risk of preeclampsia. N. Engl. J. Med. 346, 33–38. Proc. Biol. Sci. 264, 1471–1479.
Tan, C.Y., Ho, J.F., Chong, Y.S., Loganath, A., Chan, Y.H., Ravichandran, J., Xie, F., Hu, Y., Magee, L.A., Money, D.M., Patrick, D.M., Krajden, M., Thomas,
Lee, C.G., Chong, S.S., 2008. Paternal contribution of HLA-G*0106 sig- E., von Dadelszen, P., 2010. An association between cytomegalovirus
nificantly increases risk for preeclampsia in multigravid pregnancies. infection and preeclampsia: a case–control study and data synthesis.
Mol. Hum. Reprod. 14, 317–324. Acta Obstet. Gynecol. Scand. 89, 1162–1167.
Trogstad, L.I., Eskild, A., Bruu, A.L., Jeansson, S., Jenum, P.A., 2001. Is Zusterzeel, P.L., te Morsche, R., Raijmakers, M.T., Roes, E.M., Peters, W.H.,
preeclampsia an infectious disease? Acta Obstet. Gynecol. Scand. 80, Steegers, E.A., 2002. Paternal contribution to the risk for preeclampsia.
1036–1038. J. Med. Genet. 39, 44–45.

You might also like